1
|
Pokoyski C, Baars W, Windheim M, Reubold TF, Zischke J, Brinkmann A, Kay-Fedorov PC, Schwinzer R. Expression of viral CD45 ligand E3/49K on porcine cells reduces human anti-pig immune responses. Sci Rep 2023; 13:17218. [PMID: 37821577 PMCID: PMC10567836 DOI: 10.1038/s41598-023-44316-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023] Open
Abstract
Transgenic expression of protective molecules in porcine cells and tissues is a promising approach to prevent xenograft rejection. Viruses have developed various strategies to escape the host's immune system. We generated porcine B cells (B cell line L23) expressing the human adenovirus protein E3/49K or the human cytomegalovirus protein pUL11 and investigated how human T, NK and B cell responses are affected by the expression of the viral proteins. Binding studies revealed that E3/49K and pUL11 interact with CD45 on human but not porcine peripheral blood mononuclear cells. T cell proliferation in response to L23-E3/49K cells was significantly reduced and accompanied by development of an anti-inflammatory cytokine milieu (low: TNF-alpha, IFN-gamma, IL-6; high: IL-4, IL-10). Human peripheral blood mononuclear cells which had been primed for four weeks by L23-E3/49K cells included an extended population of regulatory T cells. Cytotoxicity of effector T and natural killer cells against L23 cells was significantly reduced (40 to 50%) by E3/49K expression. B cell activation and antibody production to E3/49K expressing cells was also diminished. Surprisingly, pUL11 expression showed no effects. Reduction of human anti-pig immune responses by transgenic expression of selected viral genes may be a novel approach for protection of porcine xenografts.
Collapse
Affiliation(s)
- Claudia Pokoyski
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany.
| | - Wiebke Baars
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| | - Mark Windheim
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas F Reubold
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Jasmin Zischke
- Institute of Virology, Hannover Medical School, and German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Antje Brinkmann
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| | - Penelope C Kay-Fedorov
- Institute of Virology, Hannover Medical School, and German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Reinhard Schwinzer
- Surgical Research Laboratory, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Sykes M, Sachs DH. Progress in xenotransplantation: overcoming immune barriers. Nat Rev Nephrol 2022; 18:745-761. [PMID: 36198911 DOI: 10.1038/s41581-022-00624-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
A major limitation of organ allotransplantation is the insufficient supply of donor organs. Consequently, thousands of patients die every year while waiting for a transplant. Progress in xenotransplantation that has permitted pig organ graft survivals of years in non-human primates has led to renewed excitement about the potential of this approach to alleviate the organ shortage. In 2022, the first pig-to-human heart transplant was performed on a compassionate use basis, and xenotransplantation experiments using pig kidneys in deceased human recipients provided encouraging data. Many advances in xenotransplantation have resulted from improvements in the ability to genetically modify pigs using CRISPR-Cas9 and other methodologies. Gene editing has the capacity to generate pig organs that more closely resemble those of humans and are hence more physiologically compatible and less prone to rejection. Despite such modifications, immune responses to xenografts remain powerful and multi-faceted, involving innate immune components that do not attack allografts. Thus, the induction of innate and adaptive immune tolerance to prevent rejection while preserving the capacity of the immune system to protect the recipient and the graft from infection is desirable to enable clinical xenotransplantation.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA. .,Department of Surgery, Columbia University, New York, NY, USA. .,Department of Microbiology and Immunology, Columbia University, New York, NY, USA.
| | - David H Sachs
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA. .,Department of Surgery, Columbia University, New York, NY, USA.
| |
Collapse
|
3
|
Zhou Q, Li T, Wang K, Zhang Q, Geng Z, Deng S, Cheng C, Wang Y. Current status of xenotransplantation research and the strategies for preventing xenograft rejection. Front Immunol 2022; 13:928173. [PMID: 35967435 PMCID: PMC9367636 DOI: 10.3389/fimmu.2022.928173] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Transplantation is often the last resort for end-stage organ failures, e.g., kidney, liver, heart, lung, and pancreas. The shortage of donor organs is the main limiting factor for successful transplantation in humans. Except living donations, other alternatives are needed, e.g., xenotransplantation of pig organs. However, immune rejection remains the major challenge to overcome in xenotransplantation. There are three different xenogeneic types of rejections, based on the responses and mechanisms involved. It includes hyperacute rejection (HAR), delayed xenograft rejection (DXR) and chronic rejection. DXR, sometimes involves acute humoral xenograft rejection (AHR) and cellular xenograft rejection (CXR), which cannot be strictly distinguished from each other in pathological process. In this review, we comprehensively discussed the mechanism of these immunological rejections and summarized the strategies for preventing them, such as generation of gene knock out donors by different genome editing tools and the use of immunosuppressive regimens. We also addressed organ-specific barriers and challenges needed to pave the way for clinical xenotransplantation. Taken together, this information will benefit the current immunological research in the field of xenotransplantation.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, 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
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People’s Hospital, Chengdu, China
| | - Kaiwen Wang
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Qi Zhang
- School of Medicine, University of Electronics and Technology of China, Chengdu, China
| | - Zhuowen Geng
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH, United States
- *Correspondence: Chunming Cheng, ; Yi Wang,
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China
- *Correspondence: Chunming Cheng, ; Yi Wang,
| |
Collapse
|
4
|
Zhang S, Zhou Y, Xian H, Shi Y, Liu Y, Li Z, Huang Y. Nerve regeneration in rat peripheral nerve allografts: An assessment of the role of endogenous neurotrophic factors in nerve cryopreservation and regeneration. Eur J Neurosci 2022; 55:1895-1916. [PMID: 35332602 DOI: 10.1111/ejn.15655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 11/29/2022]
Abstract
Peripheral nerve injury is a common clinical problem that often leads to significant functional impairment or even complete paralysis. Allograft has been proposed as a potential repair strategy for peripheral nerve injuries. Furthermore, peripheral nerve cryopreservation may result in nearly unlimited supply of grafts. However, the concentration of neurotrophic factors secreted by Schwann cells (SCs) in the local microenvironment after transplantation may not be sufficient for the survival of neuronal soma and axonal regeneration. Here, we investigated the effect of endogenous neurotrophic factors (ENTFs) on nerve regeneration in rats after the allograft of a cryopreserved sciatic nerve. ENTFs were highly expressed in the sciatic nerves pretreated for 14 days. Although the number of surviving cells in the sciatic nerves and their immunogenicity were low in the 14-day group after 4 weeks of cryopreservation, they continued to express high levels of ENTFs in vitro. At one week postoperation, the 14-day Allo group showed low plasma levels of interleukin-2, interferon-gamma, and tumour necrosis factor-alpha and low cellular immune response. At 20 weeks postoperation, nerve regeneration and functional recovery in the 14-day Allo group was similar to that in the fresh isograft group but better than that in the cryopreserved fresh allograft and fresh allograft groups. Thus, ENTFs were induced in vitro after pretreatment of the sciatic nerve. Following cryopreservation, the sciatic nerves with high levels of ENTFs continued to express high levels of ENTFs in vitro. The immune response after allograft was weak, which promoted recipient nerve regeneration.
Collapse
Affiliation(s)
- Song Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China.,Yubei District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ying Zhou
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Hua Xian
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Yifeng Shi
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Yunxiao Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Zijian Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China.,Nanchong Hospital of Traditional Chinese Medicine, Nanchong, China
| | - Yingru Huang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Liu Y, Liu X, Zhou S, Xu R, Hu J, Liao G, Liao J, Guo Z, Li Y, Yang S, Li S, Chen H, Guo Y, Li M, Fan L, Li L, Zhao M, Liu D. Single-Cell Profiling of Kidney Transplant Recipients With Immunosuppressive Treatment Reveals the Dynamic Immune Characteristics. Front Immunol 2021; 12:639942. [PMID: 33959124 PMCID: PMC8093626 DOI: 10.3389/fimmu.2021.639942] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/31/2021] [Indexed: 11/17/2022] Open
Abstract
Kidney transplantation is currently the first choice of treatment for various types of end-stage renal failure, but there are major limitations in the application of immunosuppressive protocols after kidney transplantation. When the dose of immunosuppressant is too low, graft rejection occurs easily, while a dose that is too high can lead to graft loss. Therefore, it is very important to explore the immune status of patients receiving immunosuppressive agents after kidney transplantation. To compare the immune status of the recipient’s whole peripheral blood before and after receipt of immunosuppressive agents, we used single-cell cytometry by time-of-flight (CyTOF) to detect the peripheral blood immune cells in five kidney transplant recipients (KTRs) from the Department of Organ Transplantation of Zhujiang Hospital of Southern Medical University before and after receiving immunosuppressive agents. Based on CyTOF analysis, we detected 363,342 live single immune cells. We found that the immune cell types of the KTRs before and after receipt of immunosuppressive agents were mainly divided into CD4+ T cells, CD8+ T cells, B cells, NK cells/γδ T cells, monocytes/macrophages, granulocytes, and dendritic cells (DCs). After further reclustering of the above cell types, it was found that the immune cell subclusters in the peripheral blood of patients underwent major changes after receipt of immunosuppressants. After receiving immunosuppressive therapy, the peripheral blood of KTRs had significantly increased levels of CD57+NK cells and significantly decreased levels of central memory CD4+ T cells, follicular helper CD4+ T cells, effector CD8+ T cells, effector memory CD8+ T cells and naive CD8+ T cells. This study used CyTOF to classify immune cells in the peripheral blood of KTRs before and after immunosuppressive treatment, further compared differences in the proportions of the main immune cell types and immune cell subgroups before and after receipt of immunosuppressants, and provided relatively accurate information for assessment and treatment strategies for KTRs.
Collapse
Affiliation(s)
- Yongguang Liu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyou Liu
- Department of Organ Transplantation, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Song Zhou
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ruiquan Xu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianmin Hu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guorong Liao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Liao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zefeng Guo
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuzhu Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siqiang Yang
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shichao Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Chen
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Guo
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Min Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lipei Fan
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liuyang Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ming Zhao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ding Liu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
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: 8.7] [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
|
7
|
Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2020; 4:4/41/eaau6298. [PMID: 31676497 DOI: 10.1126/sciimmunol.aau6298] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
Abstract
The success of organ transplantation is limited by the complications of immunosuppression, by chronic rejection, and by the insufficient organ supply, and thousands of patients die every year while waiting for a transplant. With recent progress in xenotransplantation permitting porcine organ graft survival of months or even years in nonhuman primates, there is renewed interest in its potential to alleviate the organ shortage. Many of these advances are the result of our heightened capacity to modify pigs genetically, particularly with the development of CRISPR-Cas9-based gene editing methodologies. Although this approach allows the engineering of pig organs that are less prone to rejection, the clinical application of xenotransplantation will require the ability to avoid the ravages of a multifaceted attack on the immune system while preserving the capacity to protect both the recipient and the graft from infectious microorganisms. In this review, we will discuss the potential and limitations of these modifications and how the engineering of the graft can be leveraged to alter the host immune response so that all types of immune attack are avoided.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA. .,Department of Microbiology and Immunology, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| | - David H Sachs
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| |
Collapse
|
8
|
Shu S, Ren J, Song J. Cardiac xenotransplantation: a promising way to treat advanced heart failure. Heart Fail Rev 2020; 27:71-91. [DOI: 10.1007/s10741-020-09989-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
9
|
Chen AM, Burlak C. Xenotransplantation literature update, January/February 2020. Xenotransplantation 2020; 27:e12589. [PMID: 32170808 DOI: 10.1111/xen.12589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Angela M Chen
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christopher Burlak
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
10
|
Nauman G, Borsotti C, Danzl N, Khosravi-Maharlooei M, Li HW, Chavez E, Stone S, Sykes M. Reduced positive selection of a human TCR in a swine thymus using a humanized mouse model for xenotolerance induction. Xenotransplantation 2020; 27:e12558. [PMID: 31565822 PMCID: PMC7007369 DOI: 10.1111/xen.12558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tolerance-inducing approaches to xenotransplantation would be optimal and may be necessary for long-term survival of transplanted pig organs in human patients. The ideal approach would generate donor-specific unresponsiveness to the pig organ without suppressing the patient's normal immune function. Porcine thymus transplantation has shown efficacy in promoting xenotolerance in humanized mice and large animal models. However, murine studies demonstrate that T cells selected in a swine thymus are positively selected only by swine thymic epithelial cells, and therefore, cells expressing human HLA-restricted TCRs may not be selected efficiently in a transplanted pig thymus. This may lead to suboptimal patient immune function. METHODS To assess human thymocyte selection in a pig thymus, we used a TCR transgenic humanized mouse model to study positive selection of cells expressing the MART1 TCR, a well-characterized human HLA-A2-restricted TCR, in a grafted pig thymus. RESULTS Positive selection of T cells expressing the MART1 TCR was inefficient in both a non-selecting human HLA-A2- or swine thymus compared with an HLA-A2+ thymus. Additionally, CD8 MART1 TCRbright T cells were detected in the spleens of mice transplanted with HLA-A2+ thymi but were significantly reduced in the spleens of mice transplanted with swine or HLA-A2- thymi. [Correction added on October 15, 2019, after first online publication: The missing superscript values +, -, and bright have been included in the Results section.] CONCLUSIONS: Positive selection of cells expressing a human-restricted TCR in a transplanted pig thymus is inefficient, suggesting that modifications to improve positive selection of cells expressing human-restricted TCRs in a pig thymus may be necessary to support development of a protective human T-cell pool in future patients.
Collapse
Affiliation(s)
- Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Chiara Borsotti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Nichole Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Hao-Wei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Estefania Chavez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Samantha Stone
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, USA
| |
Collapse
|
11
|
Xenotransplantation tolerance: applications for recent advances in modified swine. Curr Opin Organ Transplant 2019; 23:642-648. [PMID: 30379724 DOI: 10.1097/mot.0000000000000585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The aim of this study was to review the recent progress in xenotransplantation achieved through genetic engineering and discuss the potential of tolerance induction to overcome remaining barriers to extended xenograft survival. RECENT FINDINGS The success of life-saving allotransplantation has created a demand for organ transplantation that cannot be met by the supply of human organs. Xenotransplantation is one possible solution that would allow for a nearly unlimited supply of organs. Recent genetic engineering of swine has decreased the reactivity of preformed antibodies to some, but not all, potential human recipients. Experiments using genetically modified swine organs have now resulted in survival of life-supporting kidneys for over a year. However, the grafts show evidence of antibody-mediated rejection on histology, suggesting additional measures will be required for further extension of graft survival. Tolerance induction through mixed chimerism or thymic transplantation across xenogeneic barriers would be well suited for patients with a positive crossmatch to genetically modified swine or relatively negative crossmatches to genetically modified swine, respectively. SUMMARY This review highlights the current understanding of the immunologic processes in xenotransplantation and describes the development and application of strategies designed to overcome them from the genetic modification of the source animal to the induction of tolerance to xenografts.
Collapse
|
12
|
Kim SC, Mathews DV, Breeden CP, Higginbotham LB, Ladowski J, Martens G, Stephenson A, Farris AB, Strobert EA, Jenkins J, Walters EM, Larsen CP, Tector M, Tector AJ, Adams AB. Long-term survival of pig-to-rhesus macaque renal xenografts is dependent on CD4 T cell depletion. Am J Transplant 2019; 19:2174-2185. [PMID: 30821922 PMCID: PMC6658347 DOI: 10.1111/ajt.15329] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 01/22/2019] [Accepted: 02/04/2019] [Indexed: 01/25/2023]
Abstract
The shortage of available organs remains the greatest barrier to expanding access to transplant. Despite advances in genetic editing and immunosuppression, survival in experimental models of kidney xenotransplant has generally been limited to <100 days. We found that pretransplant selection of recipients with low titers of anti-pig antibodies significantly improved survival in a pig-to-rhesus macaque kidney transplant model (6 days vs median survival time 235 days). Immunosuppression included transient pan-T cell depletion and an anti-CD154-based maintenance regimen. Selective depletion of CD4+ T cells but not CD8+ T cells resulted in long-term survival (median survival time >400 days vs 6 days). These studies suggested that CD4+ T cells may have a more prominent role in xenograft rejection compared with CD8+ T cells. Although animals that received selective depletion of CD8+ T cells showed signs of early cellular rejection (marked CD4+ infiltrates), animals receiving selective CD4+ depletion exhibited normal biopsy results until late, when signs of chronic antibody rejection were present. In vitro study results suggested that rhesus CD4+ T cells required the presence of SLA class II to mount an effective proliferative response. The combination of low pretransplant anti-pig antibody and CD4 depletion resulted in consistent, long-term xenograft survival.
Collapse
Affiliation(s)
- SC Kim
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - DV Mathews
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - CP Breeden
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - LB Higginbotham
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - J Ladowski
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - G Martens
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - A Stephenson
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - AB Farris
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - EA Strobert
- Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - J Jenkins
- Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - EM Walters
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - CP Larsen
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia,Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - M Tector
- Comprehensive Transplant Institute, University of Alabama Birmingham School of Medicine, Birmingham, Alabama
| | - AJ Tector
- Comprehensive Transplant Institute, University of Alabama Birmingham School of Medicine, Birmingham, Alabama
| | - AB Adams
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia,Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
13
|
Sake HJ, Frenzel A, Lucas-Hahn A, Nowak-Imialek M, Hassel P, Hadeler KG, Hermann D, Becker R, Eylers H, Hein R, Baars W, Brinkmann A, Schwinzer R, Niemann H, Petersen B. Possible detrimental effects of beta-2-microglobulin knockout in pigs. Xenotransplantation 2019; 26:e12525. [PMID: 31119817 DOI: 10.1111/xen.12525] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Despite major improvements in pig-to-primate xenotransplantation, long-term survival of xenografts is still challenging. The major histocompatibility complex (MHC) class I, which is crucial in cellular immune response, is an important xenoantigen. Abrogating MHC class I expression on xenografts might be beneficial for extending graft survival beyond current limits. METHODS In this study, we employed the CRISPR/Cas9 system to target exon 2 of the porcine beta-2-microglobulin (B2M) gene to abrogate SLA class I expression on porcine cells. B2M-KO cells served as donor cells for somatic cell nuclear transfer, and cloned embryos were transferred to three recipient sows. The offspring were genotyped for mutations at the B2M locus, and blood samples were analyzed via flow cytometry for the absence of SLA class I molecules. RESULTS Pregnancies were successfully established and led to the birth of seven viable piglets. Genomic sequencing proved that all piglets carried biallelic modifications at the B2M locus leading to a frameshift, a premature stop codon, and ultimately a functional knockout. However, survival times of these animals did not exceed 4 weeks due to unexpected disease processes. CONCLUSION Here, we demonstrate the feasibility of generating SLA class I knockout pigs by targeting the porcine beta-2-microglobulin gene using the CRISPR/Cas9 system. Additionally, our findings indicate for the first time that this genetic modification might have a negative impact on the viability of the animals. These issues need to be solved to unveil the real value for xenotransplantation in the future.
Collapse
Affiliation(s)
| | - Antje Frenzel
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Andrea Lucas-Hahn
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Monika Nowak-Imialek
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Petra Hassel
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Klaus-Gerd Hadeler
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Doris Hermann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Roswitha Becker
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Heinke Eylers
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Rabea Hein
- Transplant Laboratory, Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Wiebke Baars
- Transplant Laboratory, Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Antje Brinkmann
- Transplant Laboratory, Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Reinhard Schwinzer
- Transplant Laboratory, Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Heiner Niemann
- REBIRTH/Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| |
Collapse
|
14
|
Zhao J, Lai L, Ji W, Zhou Q. Genome editing in large animals: current status and future prospects. Natl Sci Rev 2019; 6:402-420. [PMID: 34691891 PMCID: PMC8291540 DOI: 10.1093/nsr/nwz013] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
Large animals (non-human primates, livestock and dogs) are playing important roles in biomedical research, and large livestock animals serve as important sources of meat and milk. The recently developed programmable DNA nucleases have revolutionized the generation of gene-modified large animals that are used for biological and biomedical research. In this review, we briefly introduce the recent advances in nuclease-meditated gene editing tools, and we outline these editing tools' applications in human disease modeling, regenerative medicine and agriculture. Additionally, we provide perspectives regarding the challenges and prospects of the new genome editing technology.
Collapse
Affiliation(s)
- Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Liangxue Lai
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedicine Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Shanghai 200031, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
15
|
Smood B, Hara H, Schoel LJ, Cooper DKC. Genetically-engineered pigs as sources for clinical red blood cell transfusion: What pathobiological barriers need to be overcome? Blood Rev 2019; 35:7-17. [PMID: 30711308 DOI: 10.1016/j.blre.2019.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/02/2019] [Accepted: 01/25/2019] [Indexed: 12/27/2022]
Abstract
An alternative to human red blood cells (RBCs) for clinical transfusion would be advantageous, particularly in situations of massive acute blood loss (where availability and compatibility are limited) or chronic hematologic diseases requiring frequent transfusions (resulting in alloimmunization). Ideally, any alternative must be neither immunogenic nor pathogenic, but readily available, inexpensive, and physiologically effective. Pig RBCs (pRBCs) provide a promising alternative due to their several similarities with human RBCs, and our increasing ability to genetically-modify pigs to reduce cellular immunogenicity. We briefly summarize the history of xenotransfusion, the progress that has been made in recent years, and the remaining barriers. These barriers include prevention of (i) human natural antibody binding to pRBCs, (ii) their phagocytosis by macrophages, and (iii) the T cell adaptive immune response (in the absence of exogenous immunosuppressive therapy). Although techniques of genetic engineering have advanced in recent years, novel methods to introduce human transgenes into pRBCs (which do not have nuclei) will need to be developed before clinical trials can be initiated.
Collapse
Affiliation(s)
- Benjamin Smood
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leah J Schoel
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
16
|
Lee TK, Kim JM, Choi SH. Pig tracheal patchy xenotransplantation in the dog. Xenotransplantation 2018; 26:e12452. [PMID: 30117191 DOI: 10.1111/xen.12452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/08/2018] [Accepted: 07/20/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND A long-segmental tracheal lesion is difficult to repair by tracheal allotransplantation due to the lack of a well-defined blood supply for blood vessel anastomosis. The donor trachea needs to be revascularized within a well-vascularized soft tissue flap for several months to allow successful trachea allotransplantation. To date, xenotransplantation using the wild-type or genetically modified pig has been widely studied. The object of this study was to evaluate the feasibility of a small-sized (2 × 2 cm) wild-type pig tracheal patchy in a dog tracheal defect model before trying a long-segment tracheal defect model and using a genetically modified pig as a donor in dog xenotransplantation. METHOD Three healthy beagle dogs (8-9 kg) were used as recipients, and one pig (20 kg) was used as the donor. A pig cartilaginous tracheal patchy (2 × 2 cm half tube) was sutured to the tracheal resected site in each dog. Antithymocyte globulin (2.5 mg/kg infusion, D0 and 1), tacrolimus (4.5 mg/kg, twice a day for 2 months), and methylprednisolone sodium succinate (1 mg/kg, IV, for 2 days and tapering) were administered for immunosuppression. The levels IL-2 and IFN-γ in the serum were measured at D0, 7, and 28. Tracheoscopy was performed at D28, 60, and 90. The recipients were sacrificed at D90, and the expression of dog and pig genes in the graft was evaluated by PCR. Histopathological examination of the graft was conducted. RESULTS All of the dogs survived without complications during the experimental period. Their IL-2 and IFN-γ levels were significantly increased at D7 after transplantation compared to D0 and D28 (P < 0.05). The pig tracheal patchy site was open, and no stenosis was observed until D90 on tracheoscopy, when pale mucosa erosion was observed; there was also remnant suture material at D28. However, the tracheal patchy sites gradually became similar to normal mucosa at D60 and 90. The expression of pig genes was detected in the graft by PCR. Normal epithelium and CD3 cells were observed in the histological examination at D90. CONCLUSION In this study, our data suggest that the pig tracheal patchy can be successfully engrafted into the trachea of dog, although erosion of mucosa on the graft was seen at D30, in spite of the discordant species.
Collapse
Affiliation(s)
- Tae-Ki Lee
- Department of Veterinary Surgery, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Jong-Min Kim
- Xenotransplantation Research Center, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University, Seoul, Korea
| | - Seok Hwa Choi
- Department of Veterinary Surgery, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
17
|
Abstract
The growing shortage of available organs is a major problem in transplantology. Thus, new and alternative sources of organs need to be found. One promising solution could be xenotransplantation, i.e., the use of animal cells, tissues and organs. The domestic pig is the optimum donor for such transplants. However, xenogeneic transplantation from pigs to humans involves high immune incompatibility and a complex rejection process. The rapid development of genetic engineering techniques enables genome modifications in pigs that reduce the cross-species immune barrier.
Collapse
|
18
|
Smith KE, Johnson RC, Papas KK. Update on cellular encapsulation. Xenotransplantation 2018; 25:e12399. [DOI: 10.1111/xen.12399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Kate E. Smith
- Department of Physiological Sciences; University of Arizona; Tucson AZ USA
- Department of Surgery; University of Arizona; Tucson AZ USA
| | | | | |
Collapse
|
19
|
Abstract
The availability of cells, tissues and organs from a non-human species such as the pig could, at least in theory, meet the demand of organs necessary for clinical transplantation. At this stage, the important goal of getting over the first year of survival has been reported for both cellular and solid organ xenotransplantation in relevant preclinical primate models. In addition, xenotransplantation is already in the clinic as shown by the broad use of animal-derived medical devices, such as bioprosthetic heart valves and biological materials used for surgical tissue repair. At this stage, however, prior to starting a wide-scale clinical application of xenotransplantation of viable cells and organs, the important obstacle represented by the humoral immune response will need to be overcome. Likewise, the barriers posed by the activation of the innate immune system and coagulative pathway will have to be controlled. As far as xenogeneic nonviable xenografts, increasing evidence suggests that considerable immune reactions, mediated by both innate and adaptive immunity, take place and influence the long-term outcome of xenogeneic materials in patients, possibly precluding the use of bioprosthetic heart valves in young individuals. In this context, the present article provides an overview of current knowledge on the immune processes following xenotransplantation and on the possible therapeutic interventions to overcome the immunological drawbacks involved in xenotransplantation.
Collapse
Affiliation(s)
- M Vadori
- CORIT (Consortium for Research in Organ Transplantation), Via dell'Università 10, 35020 Legnaro, Padua, Italy
| | - E Cozzi
- CORIT (Consortium for Research in Organ Transplantation), Via dell'Università 10, 35020 Legnaro, Padua, Italy.,Transplant Immunology Unit, Department of Transfusion Medicine, Padua University Hospital, Via Giustiniani, 2, 35128 Padua, Italy
| |
Collapse
|
20
|
The production of multi-transgenic pigs: update and perspectives for xenotransplantation. Transgenic Res 2016; 25:361-74. [PMID: 26820415 DOI: 10.1007/s11248-016-9934-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022]
Abstract
The domestic pig shares many genetic, anatomical and physiological similarities to humans and is thus considered to be a suitable organ donor for xenotransplantation. However, prior to clinical application of porcine xenografts, three major hurdles have to be overcome: (1) various immunological rejection responses, (2) physiological incompatibilities between the porcine organ and the human recipient and (3) the risk of transmitting zoonotic pathogens from pig to humans. With the introduction of genetically engineered pigs expressing high levels of human complement regulatory proteins or lacking expression of α-Gal epitopes, the HAR can be consistently overcome. However, none of the transgenic porcine organs available to date was fully protected against the binding of anti-non-Gal xenoreactive natural antibodies. The present view is that long-term survival of xenografts after transplantation into primates requires additional modifications of the porcine genome and a specifically tailored immunosuppression regimen compliant with current clinical standards. This requires the production and characterization of multi-transgenic pigs to control HAR, AVR and DXR. The recent emergence of new sophisticated molecular tools such as Zinc-Finger nucleases, Transcription-activator like endonucleases, and the CRISPR/Cas9 system has significantly increased efficiency and precision of the production of genetically modified pigs for xenotransplantation. Several candidate genes, incl. hTM, hHO-1, hA20, CTLA4Ig, have been explored in their ability to improve long-term survival of porcine xenografts after transplantation into non-human primates. This review provides an update on the current status in the production of multi-transgenic pigs for xenotransplantation which could bring porcine xenografts closer to clinical application.
Collapse
|
21
|
Yu X, Lu L, Liu Z, Yang T, Gong X, Ning Y, Jiang Y. Brain-derived neurotrophic factor modulates immune reaction in mice with peripheral nerve xenotransplantation. Neuropsychiatr Dis Treat 2016; 12:685-94. [PMID: 27099498 PMCID: PMC4820192 DOI: 10.2147/ndt.s98387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has been demonstrated to play an important role in survival, differentiation, and neurite outgrowth for many types of neurons. This study was designed to identify the role of BDNF during peripheral nerve xenotransplantation. MATERIALS AND METHODS A peripheral nerve xenotransplantation from rats to mice was performed. Intracellular cytokines were stained for natural killer (NK) cells, natural killer T (NKT) cells, T cells, and B cells and analyzed by flow cytometry in the spleen of the recipient mouse. Serum levels of related cytokines were quantified by cytometric bead array. RESULTS Splenic NK cells significantly increased in the xenotransplanted mice (8.47±0.88×10(7) cells/mL) compared to that in the control mice (4.66±0.78×10(7) cells/mL, P=0.0003), which significantly reduced in the presence of BDNF (4.85±0.87×10(7) cells/mL, P=0.0004). In contrast, splenic NKT cell number was significantly increased in the mice with xenotransplantation plus BDNF (XT + BDNF) compared to that of control group or of mice receiving xenotransplantation only (XT only). Furthermore, the number of CD3+ T cells, CD3+CD4+ T cells, CD3+CD4- T cells, interferon-γ-producing CD3+CD4+ T cells, and interleukin (IL)-17-producing CD3+CD4+ T cells, as well as CD3-CD19+ B cells, was significantly higher in the spleen of XT only mice compared to the control mice (P<0.05), which was significantly reduced by BDNF (P<0.05). The number of IL-4-producing CD3+CD4+ T cells and CD3+CD4+CD25+Foxp3+ T cells was significantly higher in the spleen of XT + BDNF mice than that in the spleen of XT only mice (P<0.05). Serum levels of IL-6, TNF-α, interferon-γ, and IL-17 were decreased, while IL-4 and IL-10 were stimulated by BDNF following xenotransplantation. CONCLUSION BDNF reduced NK cells but increased NKT cell accumulation in the spleen of xenotransplanted mice. BDNF modulated the number of splenic T cells and its subtype cells in the mice following xenotransplantation. These findings suggest that BDNF inhibits rejection of peripheral nerve following xenotransplantation by regulating innate as well as adaptive immune reaction.
Collapse
Affiliation(s)
- Xin Yu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhigang Liu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Teng Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xu Gong
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yubo Ning
- Department of Orthopedics, Ningshi Orthopedics Hospital of Tonghua, Tonghua, People's Republic of China
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
22
|
Lin Y, Miyagi N, Byrne GW, Du Z, Kogelberg H, Gazi MH, Tazelaar HD, Wang C, McGregor CGA. A pig-to-mouse coronary artery transplantation model for investigating the pathogenicity of anti-pig antibody. Xenotransplantation 2015; 22:458-67. [PMID: 26490445 PMCID: PMC10022689 DOI: 10.1111/xen.12198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/25/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Rejection of Gal-free (GTKO) donor pig cardiac xenografts is strongly associated with vascular non-Gal antibody binding, endothelial cell (EC) injury, and activation and microvascular thrombosis. We adopted a pig-to-SCID/beige small animal transplant model to compare the pathogenicity of baboon and human anti-pig antibody. METHODS Wild-type (GT(+) ) or GTKO porcine coronary arteries (PCAs) were transplanted into the infrarenal aorta of SCID/beige mice. Three days after transplant, recipients were infused with anti-pig antibody (anti-SLA class I, an isotype control, naive or sensitized baboon serum, or naive human serum). PCAs were recovered 24 h after antibody infusion and examined using histology, immunohistochemistry, and in situ hybridization. RESULTS Dose-dependent intragraft thrombosis occurred after infusion of anti-SLA I antibody (but not isotype control) in GT(+) and GTKO PCA recipients. Naive baboon serum induced thrombosis in GT(+) grafts. Thrombosis was significantly reduced by pre-treating naive baboon serum with Gal polymer and not observed when this serum was infused to GTKO PCA recipients. Naive human serum caused dose-dependent intragraft thrombosis of GTKO PCAs. In all cases, thrombosis involved graft-specific vascular antibody and complement deposition, macrophage adherence, EC delamination, and subendothelial thrombus formation. CONCLUSIONS This study provides the first direct in vivo comparison of the pathogenicity of naive human and baboon serum. The results suggest that human preformed non-Gal antibody may have increased pathogenicity compared to baboon. This model, which showed a rejected graft histopathology similar to antibody-mediated rejection in cardiac xenotransplantation, may be useful to assess the pathogenicity of individual protein or carbohydrate specific non-Gal reactive antibodies.
Collapse
Affiliation(s)
- Yi Lin
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Cardiovascular Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Naoto Miyagi
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Guerard W Byrne
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Institute of Cardiovascular Science, University College London, London, UK
| | - Zeji Du
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Heide Kogelberg
- Institute of Cardiovascular Science, University College London, London, UK
| | | | - Henry D Tazelaar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Christopher G A McGregor
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|
23
|
Ma L, Zhang H, Hu K, Lv G, Fu Y, Ayana DA, Zhao P, Jiang Y. The imbalance between Tregs, Th17 cells and inflammatory cytokines among renal transplant recipients. BMC Immunol 2015; 16:56. [PMID: 26400627 PMCID: PMC4581081 DOI: 10.1186/s12865-015-0118-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 09/02/2015] [Indexed: 12/21/2022] Open
Abstract
Background A significant barrier to organ transplantation is the cellular rejection that occurs and mediated by antibodies, T cells, and innate immune cells. This study was aimed to determine the number of CD4+CD25+Foxp3+ Treg, CD4+IFN-γ−IL-17+ Th17, CD4+IFN-γ+IL-17− Th1 and CD4+IFN-γ+IL-17+ Th1/17 cells in renal transplant recipients (RTR). Methods Renal transplantation was performed for a total of 35 patients with end-stage renal failure. The number of CD4+CD25+Foxp3+ Treg, CD4+IFN-γ−IL-17+ Th17, CD4+IFN-γ+IL-17− Th1 and CD4+IFN-γ+IL-17+ Th1/17 cells, and the serum level of IFN-γ, TNF-α, IL-2, IL-4, IL-6, IL-10, and IL-17 were measured in pre- and post-transplant patients and 10 healthy controls (HC) using flow cytometry and Cytometric Bead Array (CBA). The association between the number of different subsets of CD4+ T-cells and clinical parameters were analyzed among the pre- and post-transplant patients, and the healthy controls. Results The number of CD4+IFN-γ−IL-17+ Th17, CD4+IFN-γ+IL-17− Th1 and CD4+IFN-γ+IL-17+ Th1/17 cells were significantly increased in patients with End-Stage Renal Failure (ESRF) compared to the HC. Stratification analysis indicated that AMR (Acute antibody mediated acute rejection), AR (acute rejection) and CR (chronic rejection) groups displayed greater number of CD4+IFN-γ−IL-17+ Th17, CD4+IFN-γ+IL-17− Th1 and CD4+IFN-γ+IL-17+ Th1/17 cells as well as high level of serum IL-2, IFN-γ, TNF-α and IL-17. But, the AMR, AR and CR groups have shown lower level of CD4+CD25+Foxp3+ T cells and serum IL-10 compared to transplant stable (TS) patients. Moreover, the number of Tregs were negatively correlated with the number of Th17 cells in RTR patients. The number of Tregs and Th17 cells were positively correlated with the eGFR and serum creatinine values, respectively. Conclusion The imbalance between different types of CD4+ T cells and dysregulated inflammatory cytokines may contribute towards renal transplantation rejection.
Collapse
Affiliation(s)
- Liang Ma
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China. .,Department of Gastroenterology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, 213003, China.
| | - Huimao Zhang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Kebang Hu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Guoyue Lv
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yaowen Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| | | | - Pingwei Zhao
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
24
|
Griesemer A, Yamada K, Sykes M. Xenotransplantation: immunological hurdles and progress toward tolerance. Immunol Rev 2015; 258:241-58. [PMID: 24517437 DOI: 10.1111/imr.12152] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The discrepancy between organ need and organ availability represents one of the major limitations in the field of transplantation. One possible solution to this problem is xenotransplantation. Research in this field has identified several obstacles that have so far prevented the successful development of clinical xenotransplantation protocols. The main immunologic barriers include strong T-cell and B-cell responses to solid organ and cellular xenografts. In addition, components of the innate immune system can mediate xenograft rejection. Here, we review these immunologic and physiologic barriers and describe some of the strategies that we and others have developed to overcome them. We also describe the development of two strategies to induce tolerance across the xenogeneic barrier, namely thymus transplantation and mixed chimerism, from their inception in rodent models through their current progress in preclinical large animal models. We believe that the addition of further beneficial transgenes to Gal knockout swine, combined with new therapies such as Treg administration, will allow for successful clinical application of xenotransplantation.
Collapse
Affiliation(s)
- Adam Griesemer
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | | | |
Collapse
|
25
|
Byrne GW, Du Z, Stalboerger P, Kogelberg H, McGregor CGA. Cloning and expression of porcine β1,4 N-acetylgalactosaminyl transferase encoding a new xenoreactive antigen. Xenotransplantation 2014; 21:543-54. [PMID: 25176027 PMCID: PMC4262693 DOI: 10.1111/xen.12124] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/28/2014] [Indexed: 01/03/2023]
Abstract
Background Xenograft rejection of pigs organs with an engineered mutation in the GGTA-1 gene (GTKO) remains a predominantly antibody mediated process which is directed to a variety of non-Gal protein and carbohydrate antigens. We previously used an expression library screening strategy to identify six porcine endothelial cell cDNAs which encode pig antigens that bind to IgG induced after pig-to-primate cardiac xenotransplantation. One of these gene products was a glycosyltransferase with homology to the bovine β1,4 N-acetylgalactosaminyltransferase (B4GALNT2). We now characterize the porcine B4GALNT2 gene sequence, genomic organization, expression, and functional significance. Methods The porcine B4GALNT2 cDNA was recovered from the original library isolate, subcloned, sequenced, and used to identify a bacterial artificial chromosome (BAC) containing the entire B4GALNT2 locus from the Children's Hospital Oakland Research Institute BACPAC Resource Centre (#AC173453). PCR primers were designed to map the intron/exon genomic organization in the BAC clone. A stable human embryonic kidney (HEK) cell line expressing porcine B4GALNT2 (HEK-B4T) was produced. Expression of porcine B4GALNT2 in HEK-B4T cells was characterized by immune staining and siRNA transfection. The effects of B4GALNT2 expression in HEK-B4T cells was measured by flow cytometry and complement mediated lysis. Antibody binding to HEK and HEK-B4T cells was used to detect an induced antibody response to the B4GALNT2 produced glycan and the results were compared to GTKO PAEC specific non-Gal antibody induction. Expression of porcine B4GALNT2 in pig cells and tissues was measured by qualitative and quantitative real time reverse transcriptase PCR and by Dolichos biflorus agglutinin (DBA) tissue staining. Results The porcine B4GALNT2 gene shares a conserved genomic organization and encodes an open reading frame with 76 and 70% amino acid identity to the human and murine B4GALNT2 genes, respectively. The B4GALNT2 gene is expressed in porcine endothelial cells and shows a broadly distributed expression pattern. Expression of porcine B4GALNT2 in human HEK cells (HEK-B4T) results in increased binding of antibody to the B4GALNT2 enzyme, and increased reactivity with anti-Sda and DBA. HEK-B4T cells show increased sensitivity to complement mediated lysis when challenged with serum from primates after pig to primate cardiac xenotransplantation. In GTKO and GTKO:CD55 cardiac xenotransplantation recipients there is a significant correlation between the induction of a non-Gal antibody, measured using GTKO PAECs, and the induction of antibodies which preferentially bind to HEK-B4T cells. Conclusion The functional isolation of the porcine B4GALNT2 gene from a PAEC expression library, the pattern of B4GALNT2 gene expression and its sensitization of HEK-B4T cells to antibody binding and complement mediated lysis indicates that the enzymatic activity of porcine B4GALNT2 produces a new immunogenic non-Gal glycan which contributes in part to the non-Gal immune response detected after pig-to-baboon cardiac xenotransplantation.
Collapse
Affiliation(s)
- Guerard W Byrne
- Department of Surgery, Mayo Clinic, Rochester, MN, USA; Institute of Cardiovascular Science, University College London, London, UK
| | | | | | | | | |
Collapse
|
26
|
Azimzadeh AM, Byrne GW, Ezzelarab M, Welty E, Braileanu G, Cheng X, Robson SC, McGregor CGA, Cooper DKC, Pierson RN. Development of a consensus protocol to quantify primate anti-non-Gal xenoreactive antibodies using pig aortic endothelial cells. Xenotransplantation 2014; 21:555-66. [PMID: 25176173 DOI: 10.1111/xen.12125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 05/30/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Scientists working in the field of xenotransplantation do not employ a uniform method to measure and report natural and induced antibody responses to non-Galα(1,3)Gal (non-Gal) epitopes. Such humoral responses are thought to be particularly pathogenic after transplantation of vascularized GalTKO pig organs and having a more uniform assay and reporting format would greatly facilitate comparisons between laboratories. METHODS Flow cytometry allows examination of antibody reactivity to intact antigens in their natural location and conformation on cell membranes. We have established a simple and reproducible flow cytometric assay to detect antibodies specific for non-Gal pig antigens using primary porcine aortic endothelial cells (pAECs) and cell culture-adapted pAEC cell lines generated from wild type and α1,3galactosyl transferase knockout (GalTKO) swine. RESULTS The consensus protocol we propose here is based on procedures routinely used in four xenotransplantation centers and was independently evaluated at three sites using shared cells and serum samples. Our observation support use of the cell culture-adapted GalTKO pAEC KO:15502 cells as a routine method to determine the reactivity of anti-non-Gal antibodies in human and baboon serum. CONCLUSIONS We have developed an assay that allows the detection of natural and induced non-Gal xenoreactive antibodies present in human or baboon serum in a reliable and consistent manner. This consensus assay and format for reporting the data should be accessible to laboratories and will be useful for assessing experimental results between multiple research centers. Adopting this assay and format for reporting the data should facilitate the detection, monitoring, and detailed characterization of non-Gal antibody responses.
Collapse
Affiliation(s)
- Agnes M Azimzadeh
- Division of Cardiac Surgery, University of Maryland and VAMC Baltimore, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cooper DKC, Satyananda V, Ekser B, van der Windt DJ, Hara H, Ezzelarab MB, Schuurman HJ. Progress in pig-to-non-human primate transplantation models (1998-2013): a comprehensive review of the literature. Xenotransplantation 2014; 21:397-419. [PMID: 25176336 DOI: 10.1111/xen.12127] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/03/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pig-to-non-human primate model is the standard choice for in vivo studies of organ and cell xenotransplantation. In 1998, Lambrigts and his colleagues surveyed the entire world literature and reported all experimental studies in this model. With the increasing number of genetically engineered pigs that have become available during the past few years, this model is being utilized ever more frequently. METHODS We have now reviewed the literature again and have compiled the data we have been able to find for the period January 1, 1998 to December 31, 2013, a period of 16 yr. RESULTS The data are presented for transplants of the heart (heterotopic and orthotopic), kidney, liver, lung, islets, neuronal cells, hepatocytes, corneas, artery patches, and skin. Heart, kidney, and, particularly, islet xenograft survival have increased significantly since 1998. DISCUSSION The reasons for this are briefly discussed. A comment on the limitations of the model has been made, particularly with regard to those that will affect progression of xenotransplantation toward the clinic.
Collapse
Affiliation(s)
- David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Vadori M, Cozzi E. Immunological challenges and therapies in xenotransplantation. Cold Spring Harb Perspect Med 2014; 4:a015578. [PMID: 24616201 DOI: 10.1101/cshperspect.a015578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Xenotransplantation, or the transplantation of cells, tissues, or organs between different species, was proposed a long time ago as a possible solution to the worldwide shortage of human organs and tissues for transplantation. In this setting, the pig is currently seen as the most likely candidate species. In the last decade, progress in this field has been remarkable and includes a better insight into the immunological mechanisms underlying the rejection process. Several immunological hurdles nonetheless remain, such as the strong antibody-mediated and innate or adaptive cellular immune responses linked to coagulation derangements, precluding indefinite xenograft survival. This article reviews our current understanding of the immunological mechanisms involved in xenograft rejection and the potential strategies that may enable xenotransplantation to become a clinical reality in the not-too-distant future.
Collapse
Affiliation(s)
- Marta Vadori
- CORIT (Consortium for Research in Organ Transplantation), Legnaro, 35020 Padua, Italy
| | | |
Collapse
|
29
|
Lee IK, Son YM, Ju YJ, Song SK, Gu M, Song KD, Lee HC, Woo JS, Seol JG, Park SM, Han SH, Yun CH. Survival of porcine fibroblasts enhanced by human FasL and dexamethasone-treated human dendritic cells in vitro. Transpl Immunol 2014; 30:99-106. [PMID: 24518159 DOI: 10.1016/j.trim.2014.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 01/28/2014] [Accepted: 01/28/2014] [Indexed: 01/08/2023]
Abstract
Cell-mediated and acute vascular rejections remain to be one of the primary hurdles to achieve successful xenotransplantation. Fas ligand is known to be an important molecule for the formation of 'immune-privileged' condition and dendritic cells treated with dexamethasone (Dex-DCs) acting like tolerogenic DCs (tDCs) which are known to protect transplanted cells and organs from unwanted immune responses. The present study investigated the possibility that porcine fibroblasts expressing human Fas ligand (PhF) together with human Dex-DCs could induce prolonged survival of porcine fibroblasts in vitro. PhF was collected from an ear of human Fas ligand transgenic porcine and cell-line was established by MGEM Inc. PhF labeled with CFSE co-cultured with human peripheral blood mononuclear cells (hPBMCs) were examined with respect to induction of tolerance and cell death when co-cultured with Dex-DCs for 3days. PhF induced the apoptosis in hPBMCs, especially CD4(+) T cells. Dex-DCs showed significant (P<0.05) reduction on the expression of CD80, CD86 and MHC class I/II, and the secretion of IL-12p70, TNF-α and IL-10, but increase of latency-associated peptide (LAP). Survival of PhF was significantly higher than that of WT and it was increased in the presence of Dex-DCs when compared to the other DCs (i.e.,DCs, LPS-treated DCs and LPS/Dex-treated DCs) in vitro. Survival of PhF did not change by co-culture with Dex-DCs due to apoptotic cell death of Dex-DCs. Dex-DCs reduced the death of porcine fibroblasts and, at the same time, PhF induced the apoptosis from hPBMCs, but it was not synergistic.
Collapse
Affiliation(s)
- In Kyu Lee
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Min Son
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Jun Ju
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sun Kwang Song
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Minjung Gu
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; WCU Biomodulation major and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Ki-Duk Song
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; WCU Biomodulation major and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Hwi-Cheul Lee
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Suwon, Republic of Korea
| | - Jae-Seok Woo
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Suwon, Republic of Korea
| | | | - Sung Moo Park
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; WCU Biomodulation major and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology & Immunology, DRI, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; WCU Biomodulation major and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Abstract
The shortage of human organs for transplantation has focused research on the possibility of transplanting pig organs into humans. Many factors contribute to the failure of a pig organ graft in a primate. A rapid innate immune response (natural anti-pig antibody, complement activation, and an innate cellular response; e.g., neutrophils, monocytes, macrophages, and natural killer cells) is followed by an adaptive immune response, although T-cell infiltration of the graft has rarely been reported. Other factors (e.g., coagulation dysregulation and inflammation) appear to play a significantly greater role than in allotransplantation. The immune responses to a pig xenograft cannot therefore be controlled simply by suppression of T-cell activity. Before xenotransplantation can be introduced successfully into the clinic, the problems of the innate, coagulopathic, and inflammatory responses will have to be overcome, most likely by the transplantation of organs from genetically engineered pigs. Many of the genetic manipulations aimed at protecting against these responses also reduce the adaptive response. The T-cell and elicited antibody responses can be prevented by the biological and/or pharmacologic agents currently available, in particular, by costimulation blockade-based regimens. The exogenous immunosuppressive regimen may be significantly reduced by the presence of a graft from a pig transgenic for a mutant (human) class II transactivator gene, resulting in down-regulation of swine leukocyte antigen class II expression, or from a pig with "local" vascular endothelial cell expression of an immunosuppressive gene (e.g., CTLA4-Ig). The immunomodulatory efficacy of regulatory T cells or mesenchymal stromal cells has been demonstrated in vitro but not yet in vivo.
Collapse
|
31
|
Hara H, Witt W, Crossley T, Long C, Isse K, Fan L, Phelps CJ, Ayares D, Cooper DKC, Dai Y, Starzl TE. Human dominant-negative class II transactivator transgenic pigs - effect on the human anti-pig T-cell immune response and immune status. Immunology 2013; 140:39-46. [PMID: 23566228 DOI: 10.1111/imm.12107] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 03/26/2013] [Accepted: 04/03/2013] [Indexed: 12/13/2022] Open
Abstract
Swine leucocyte antigen (SLA) class II molecules on porcine (p) cells play a crucial role in xenotransplantation as activators of recipient human CD4(+) T cells. A human dominant-negative mutant class II transactivator (CIITA-DN) transgene under a CAG promoter with an endothelium-specific Tie2 enhancer was constructed. CIITA-DN transgenic pigs were produced by nuclear transfer/embryo transfer. CIITA-DN pig cells were evaluated for expression of SLA class II with/without activation, and the human CD4(+) T-cell response to cells from CIITA-DN and wild-type (WT) pigs was compared. Lymphocyte subset numbers and T-cell function in CIITA-DN pigs were compared with those in WT pigs. The expression of SLA class II on antigen-presenting cells from CIITA-DN pigs was significantly reduced (40-50% reduction compared with WT; P < 0·01), and was completely suppressed on aortic endothelial cells (AECs) even after activation (100% suppression; P < 0·01). The human CD4(+) T-cell response to CIITA-DN pAECs was significantly weaker than to WT pAECs (60-80% suppression; P < 0·01). Although there was a significantly lower frequency of CD4(+) cells in the PBMCs from CIITA-DN (20%) than from WT (30%) pigs (P < 0·01), T-cell proliferation was similar, suggesting no significant immunological compromise. Organs and cells from CIITA-DN pigs should be partially protected from the human cellular immune response.
Collapse
Affiliation(s)
- Hidetaka Hara
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Li W, Jia Z, Zhang S, Lin X, Yang R, He Q, Ruan D. The cellular immune mechanism after transfer of chemically extracted acellular nerve xenografts. PLoS One 2013; 8:e68806. [PMID: 23874771 PMCID: PMC3714289 DOI: 10.1371/journal.pone.0068806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/28/2013] [Indexed: 11/19/2022] Open
Abstract
Severe peripheral nerve defect by injuries causing functional loss require nerve grafting. Autograft has limitations for clinical use because it results in the creation of a new nerve injury and the generation of donor site morbidity. Based on these limitations, nerve allografts and xenografts provide a readily accessible alternative strategy. The aim of the present study was to observe the immune mechanism underlying the rejection of chemically extracted acellular nerve xenografts, and further evaluate immunogenicity of chemically treated acellular nerve grafts for clinical applications. A total of 160 BALB/c mice were randomly divided into a negative contrast group (NC, 40 mice), a fresh autograft group (AG, 40 mice), a fresh xenogeneic nerve group (FXN, 40 mice) and a chemically extracted acellular xenogeneic nerve group (CEXN, 40 mice). Various types of nerve grafts were implanted into the thigh muscle of BALB/C mice in the corresponding groups. At 3, 7, 14 and 28 days post-operation, the mice (10 mice from each group) were sacrificed and their spleens were extracted. The spleens were ground into paste. The erythrocytes and other cells were lysed using distilled water and the T lymphocytes were collected. Fluorescein isothiocyanate (FITC) -labeled monoclonal antibodies (CD3, CD4, CD8, CD25, IL-2, IFN-γ and TNF-α) were then added to the solution. The Fluorescence Activated Cell Sorting (FACS) was used to determine the positivity rate of the cells combined with the monoclonal antibodies above. No significant statistical differences were observed between the CEXN, NC and AG groups, so that no obvious immune rejections were observed among the chemically extracted acellular nerve xenografts.
Collapse
Affiliation(s)
- Wei Li
- Department of Orthopaedics, Navy General Hospital, Beijing, China.
| | | | | | | | | | | | | |
Collapse
|
33
|
Perry DL, Bollinger L, L.White G. The Baboon (Papio spp.) as a model of human Ebola virus infection. Viruses 2012; 4:2400-16. [PMID: 23202470 PMCID: PMC3497058 DOI: 10.3390/v4102400] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 10/17/2012] [Accepted: 10/17/2012] [Indexed: 01/09/2023] Open
Abstract
Baboons are susceptible to natural Ebola virus (EBOV) infection and share 96% genetic homology with humans. Despite these characteristics, baboons have rarely been utilized as experimental models of human EBOV infection to evaluate the efficacy of prophylactics and therapeutics in the United States. This review will summarize what is known about the pathogenesis of EBOV infection in baboons compared to EBOV infection in humans and other Old World nonhuman primates. In addition, we will discuss how closely the baboon model recapitulates human EBOV infection. We will also review some of the housing requirements and behavioral attributes of baboons compared to other Old World nonhuman primates. Due to the lack of data available on the pathogenesis of Marburg virus (MARV) infection in baboons, discussion of the pathogenesis of MARV infection in baboons will be limited.
Collapse
Affiliation(s)
- Donna L. Perry
- Integrated Research Facility, Division of Clinical Research, NIAID, NIH, Frederick, MD, USA;
| | - Laura Bollinger
- Integrated Research Facility, Division of Clinical Research, NIAID, NIH, Frederick, MD, USA;
| | - Gary L.White
- Department of Pathology, University of Oklahoma Baboon Research Resource, University of Oklahoma, Ft. Reno Science Park, OK, USA;
| |
Collapse
|
34
|
Abstract
Cellular studies are essential in the xenotransplantation field in order to investigate the cellular immune responses triggered by xenogeneic cells and identify the key molecules involved. A series of functional studies can be conducted with this purpose that include treatment with proinflammatory cytokines and xenogeneic cell-based assays that put together pig cells and human leukocytes such as monocytes, NK cells, and T cells. The choice of the pig cell type is critical to appropriately model the transplant setting of interest. Thus, pig endothelial cells are commonly used for studying the rejection process of vascularized organs. Treatment with cytokines allows studying the regulation of adhesion, costimulatory molecules, and receptors involved in triggering the immune response in an attempt to reproduce the more complex in vivo situation. The adhesion assays are used to determine the capacity of human leukocytes to adhere to porcine cells under various conditions. Furthermore, we describe coculture, costimulatory, and cytotoxicity assays for investigating the cellular and molecular mechanisms that take place during the xenogeneic immune response.
Collapse
|
35
|
Yu X, Jiang Y, Lu L, Gong X, Sun X, Xuan Z, Lu L. A crucial role of IL-17 and IFN-γ during acute rejection of peripheral nerve xenotransplantation in mice. PLoS One 2012; 7:e34419. [PMID: 22479627 PMCID: PMC3316676 DOI: 10.1371/journal.pone.0034419] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 02/28/2012] [Indexed: 11/23/2022] Open
Abstract
Nerve injuries causing segmental loss require nerve grafting. However, autografts and allografts have limitations for clinical use. Peripheral nerve xenotransplantation has become an area of great interest in clinical surgery research as an alternative graft strategy. However, xenotransplant rejection is severe with cellular immunity, and Th1 cells play an important role in the process. To better understand the process of rejection, we used peripheral nerve xenografts from rats to mice and found that mononuclear cells expressing IFN-γ and IL-17 infiltrated around the grafts, and IFN-γ and IL-17 producing CD4+ and CD8+ T cells increased during the process of acute rejection. The changes of IL-4 level had no significant difference between xenotransplanted group and sham control group. The rejection of xenograft was significantly prevented after the treatment of IL-17 and IFN-γ neutralizing antibodies. These data suggest that Th17 cells contribute to the acute rejection process of peripheral nerve xenotransplant in addition to Th1 cells.
Collapse
Affiliation(s)
- Xin Yu
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| | - Yanfang Jiang
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Lu Lu
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| | - Xu Gong
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| | - Xiguang Sun
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| | - Zhaopeng Xuan
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| | - Laijin Lu
- Department of Hand Surgery, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
36
|
Koshika T, Phelps C, Fang J, Lee SE, Fujita M, Ayares D, Cooper DKC, Hara H. Relative efficiency of porcine and human cytotoxic T-lymphocyte antigen 4 immunoglobulin in inhibiting human CD4+ T-cell responses co-stimulated by porcine and human B7 molecules. Immunology 2012; 134:386-97. [PMID: 22043861 DOI: 10.1111/j.1365-2567.2011.03496.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
α1,3-Galactosyltransferase gene-knockout pigs transgenic for porcine cytotoxic T-lymphocyte antigen 4 immunoglobulin (pCTLA4-Ig) have been produced to reduce T-cell-mediated rejection following xenotransplantation. The level of soluble pCTLA4-Ig in their blood was greatly in excess of the therapeutic level in patients, rendering the pigs immune-incompetent. Soluble pCTLA4-Ig produced by these transgenic pigs was evaluated for binding to porcine and human (h) B7 molecules, and for its inhibitory effect on allogeneic and xenogeneic human T-cell responses. Porcine CTLA4-Ig-expressing peripheral blood mononuclear cells (PBMCs) and aortic endothelial cells (AECs) were evaluated for their direct inhibitory effect on hCD4+ T-cell responses. Soluble pCTLA4-Ig and purified hCTLA4-Ig showed similar binding to pB7 molecules, but pCTLA4-Ig showed significantly less binding to hB7 molecules. The pCTLA4-Ig and hCTLA4-Ig inhibited the response of hCD4+ T cells to pAECs equally, but pCTLA4-Ig was less successful in inhibiting the human allogeneic response. The hCD4+ T-cell response to PBMCs from pCTLA4-Ig pigs was significantly lower than that of non-pCTLA4-Ig pigs. Although pCTLA4-Ig was detected in the cytoplasm of pCTLA4-Ig-expressing pAECs, only a minimal level of soluble pCTLA4-Ig was detected in the supernatant during culture, and pCTLA4-Ig-expressing pAECs did not inhibit the xenogeneic direct human T-cell response. High-level tissue-specific production of pCTLA4-Ig may be required for sufficient immunosuppression for organ or cell (e.g., islets) transplantation.
Collapse
Affiliation(s)
- Tadatsura Koshika
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Identification of new carbohydrate and membrane protein antigens in cardiac xenotransplantation. Transplantation 2011; 91:287-92. [PMID: 21119562 PMCID: PMC10022691 DOI: 10.1097/tp.0b013e318203c27d] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND α1,3-Galactosyltransferase gene knockout (GTKO) pigs reduced the significance of antibody to galactose alpha 1,3-galactose (Gal) antigens but did not eliminate delayed xenograft rejection (DXR). We hypothesize that DXR of GTKO organs results from an antibody response to a limited number of non-Gal endothelial cell (EC) membrane antigens. In this study, we screened a retrovirus expression library to identify EC membrane antigens detected after cardiac xenotransplantation. METHODS Expression libraries were made from GT:CD46 and GTKO porcine aortic ECs. Viral stocks were used to infect human embryonic kidney cells (HEK) that were selected by flow cytometry for IgG binding from sensitized cardiac heterotopic xenograft recipients. After three to seven rounds of selection, individual clones were assessed for non-Gal IgG binding. The porcine complementary DNA was recovered by polymerase chain reaction amplification, sequenced, and identified by homology comparisons. RESULTS A total of 199 and 317 clones were analyzed from GT:CD46 and GTKO porcine aortic EC complementary DNA libraries, respectively. Sequence analysis identified porcine CD9, CD46, CD59, and the EC protein C receptor. We also identified porcine annexin A2 and a glycosyltransferase with homology to the human β1,4 N-acetylgalactosaminyl transferase 2 gene. CONCLUSION The identified proteins include key EC functions and suggest that non-Gal antibody responses may compromise EC functions and thereby contribute to DXR. Recovery of the porcine β1,4 N-acetylgalactosaminyl transferase 2 suggests that an antibody response to a SD-like carbohydrate may represent a new carbohydrate moiety involved in xenotransplantation. The identification of these porcine gene products may lead to further donor modification to enhance resistance to DXR and further reduce the level of xenograft antigenicity.
Collapse
|
38
|
Chae JI, Shim JH, Lee KS, Cho YS, Lee KS, Yoon DY, Kim SH, Chung HM, Koo DB, Park CS, Lee DS, Myung PK. Downregulation of immune response by the human cytokines Interleukin-32alpha and beta in cell-mediated rejection. Cell Immunol 2010; 264:47-53. [PMID: 20541181 DOI: 10.1016/j.cellimm.2010.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 04/21/2010] [Accepted: 04/26/2010] [Indexed: 12/30/2022]
Abstract
Xenotransplantation of porcine organs has the potential to help overcome the severe shortage of human tissues and organs available for human transplantation. However, numerous hurdles such as immune-mediated xenograft rejection remain before clinical xenotransplantation. In this study, we elucidated the role of human TNF-alpha-inducing factor, Interleukin-32 (IL-32), in porcine kidney cells (PK-15) during cell-mediated rejection by examining host cell responses. CD8+ and CD4+ T cells numbers were reduced in the lymph nodes of PK-15/IL-32beta injected mice. CD3+ Tcells were in mice injected with control cells but PK-15/IL-32alpha- and PK-15/IL-32beta-injected cell numbers were lower in lymph nodes than un transfected controls. In Mixed lymphocyte reaction cultures, the rates of cell proliferation were increased in both PK-15/IL-32alpha- and PK-15/IL-32beta-injected groups compared to the untransfected control groups. The Stable porcine PK-15 cells expression IL-32alpha and IL-32beta inhibited cytotoxic T lymphocyte (CTLs) after cellular xenograft. Our results suggest that human IL-32alpha and IL-32beta regulates on xenograft rejection in cellular xenotransplantation.
Collapse
Affiliation(s)
- Jung Il Chae
- Department of Oral Pharmacology, School of Dentistry, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Le Bas-Bernardet S, Blancho G. Current cellular immunological hurdles in pig-to-primate xenotransplantation. Transpl Immunol 2009; 21:60-4. [DOI: 10.1016/j.trim.2008.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 10/09/2008] [Indexed: 12/13/2022]
|
40
|
Vadori M, Seveso M, Besenzon F, Bosio E, Tognato E, Fante F, Boldrin M, Gavasso S, Ravarotto L, Mann BE, Simioni P, Ancona E, Motterlini R, Cozzi E. In vitro and in vivo effects of the carbon monoxide-releasing molecule, CORM-3, in the xenogeneic pig-to-primate context. Xenotransplantation 2009; 16:99-114. [PMID: 19392725 DOI: 10.1111/j.1399-3089.2009.00521.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Carbon monoxide (CO) interferes with inflammatory and apoptotic processes associated with ischemia-reperfusion injury and graft rejection. Here, the in vitro effects of carbon monoxide releasing molecule-3 (CORM-3), a novel water-soluble carbonyl CO carrier, have been investigated on porcine aortic endothelial cells (PAEC) and primate peripheral blood mononuclear cells (PBMC). Furthermore, the pharmacodynamics and pharmacotolerance of CORM-3 after administration of single and multiple doses in the primate have been assessed in view of its potential application in pig-to-primate xenotransplantation models. METHODS For in vitro studies, PAEC and primate PBMC were exposed for 24, 48 and 72 h to CORM-3 (20 to 1000 microm) and viability was measured using an MTS assay. PAEC and primate PBMC proliferation after exposure to CORM-3 was assessed by CFSE labelling. Proliferation of primate PBMC against irradiated pig lymphocytes was also assessed. Tumor necrosis factor alpha (TNF-alpha) production and Caspase-3 and -7 activity in Concanavalin A (conA)-stimulated primate PBMC were measured following treatment with CORM-3. In vivo, CORM-3 was administered i.v. to cynomolgus monkeys at 4 mg/kg, as single or multiple doses for up to 30 days. The effect of CORM-3 was evaluated by the assessment of production of TNF-alpha and interleukin 1beta following PBMC stimulation with LPS by species-specific ELISA. Complete hematologic and biochemical analyses were routinely performed in treated primates. RESULTS At concentrations <500 microm, CORM-3 did not alter the viability of PAEC or primate PBMC cultures in vitro, nor did it induce significant levels of apoptosis or necrosis. Interestingly, at concentrations of 300 and 500 microm, significant PAEC proliferation was observed, whilst concentrations > or =50 microm inhibited conA-activated primate lymphocyte proliferation (IC(50) of 345.8 +/- 51.9 microm) and the primate xenogeneic response against pig PBMC. Such responses were demonstrated to be CO-dependent. In addition, CORM-3 significantly inhibited caspase-3 and -7 activity at concentrations between 200 and 500 microm and caused a significant reduction in TNF-alpha production (IC(50) 332.8 +/- 33.9 microm). In vivo, following the administration of multiple doses, TNF-alpha production was significantly reduced in comparison to pre-treatment responses, with decreased levels maintained throughout the study. Moreover, a slight and transient increase in transaminases and bilirubin was observed in animals exposed to multiple doses of CORM-3. CONCLUSIONS These studies suggest that CORM-3 has anti-inflammatory and immunomodulatory properties in primates that may result in clinical benefit to allo- and xenografted organs.
Collapse
Affiliation(s)
- Marta Vadori
- Consorzio per la Ricerca sul Trapianto d'Organi, University of Padua, Via dei Giustiniani 2, Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW To review the latest development in cardiac xenotransplantation in small and large animal models and related in-vitro studies. RECENT FINDINGS With the recent introduction of alpha1,3-galactosyltransferase gene-knockout (GT-KO) pig organs for xenotransplantation, improved cardiac graft survival has been obtained. However, this experience has demonstrated the importance of pig antigens other than Galalpha1,3Gal (Gal) antigens (so-called nonGal antigens) as targets for primate anti-pig antibodies. Several in-vitro studies have confirmed that, although the incidence and levels of anti-nonGal antibodies in nonhuman primates and humans are significantly less when compared with total anti-pig antibodies (i.e., anti-Gal + anti-nonGal), they can result in complement-mediated lysis of GT-KO pig cells. More recently, it has been demonstrated that regulatory T cells suppress the cellular xenogeneic response, thus potentially preventing or reducing T-cell-mediated rejection. The importance of thrombotic microangiopathy as a feature of the immune/inflammatory response and incompatibilities between the coagulation-anticoagulation systems of pig and primate are receiving increasing attention. Development of GT-KO pigs transgenic for one or more 'antithrombotic' genes, for example, CD39 or tissue factor pathway inhibitor, may contribute to overcoming these problems. SUMMARY Although GT-KO pigs have provided an advance over wild-type pigs as a source of organs for transplantation into primates, further genetic modification of GT-KO pigs is required to overcome the remaining immune barriers before a clinical trial of cardiac xenotransplantation can be contemplated.
Collapse
|
42
|
Byrne GW, Stalboerger PG, Davila E, Heppelmann CJ, Gazi MH, McGregor HCJ, LaBreche PT, Davies WR, Rao VP, Oi K, Tazelaar HD, Logan JS, McGregor CGA. Proteomic identification of non-Gal antibody targets after pig-to-primate cardiac xenotransplantation. Xenotransplantation 2009; 15:268-76. [PMID: 18957049 DOI: 10.1111/j.1399-3089.2008.00480.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Experience with non-antigenic galactose alpha1,3 galactose (alphaGal) polymers and development of alphaGal deficient pigs has reduced or eliminated the significance of this antigen in xenograft rejection. Despite these advances, delayed xenograft rejection (DXR) continues to occur most likely due to antibody responses to non-Gal endothelial cell (EC) antigens. METHODS To gauge the diversity of the non-Gal antibody response we used antibody derived from CD46 transgenic heterotopic cardiac xenografts performed without T-cell immunosuppression, Group A (n = 4) and Gal knockout (GT-KO) heart transplants under tacrolimus and sirolimus immunosuppression, Group B (n = 8). Non-Gal antibody was measured by flow cytometry and by western blots using GT-KO EC membrane antigens. A nanoLC/MS/MS analysis of proteins recovered from 2D gels was used to identify target antigens. RESULTS Group A recipients exhibited a mixed cellular and humoral rejection. Group B recipients mainly exhibited classical DXR. Western blot analysis showed a non-Gal antibody response induced by GT+ and GT-KO hearts to an overlapping set of pig aortic EC membrane antigens. Proteomic analysis identified 14 potential target antigens but failed to define several immunodominant targets. CONCLUSIONS These experiments indicate that the non-Gal antibody response is directed to a number of stress response and inflammation related pig EC antigens and a few undefined targets. Further analysis of these antibody specificities using alternative methods is required to more fully define the repertoire of non-Gal antibody responses.
Collapse
Affiliation(s)
- Guerard W Byrne
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Park CG, Kim JS, Shin JS, Kim YH, Kim SJ. Current Status and Future Perspectives of Xenotransplantation. ACTA ACUST UNITED AC 2009. [DOI: 10.4285/jkstn.2009.23.3.203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Sik Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Jun-Seop Shin
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Yong-Hee Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Joon Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Hisashi Y, Yamada K, Kuwaki K, Tseng YL, Dor FJMF, Houser SL, Robson SC, Schuurman HJ, Cooper DKC, Sachs DH, Colvin RB, Shimizu A. Rejection of cardiac xenografts transplanted from alpha1,3-galactosyltransferase gene-knockout (GalT-KO) pigs to baboons. Am J Transplant 2008; 8:2516-26. [PMID: 19032222 PMCID: PMC2836186 DOI: 10.1111/j.1600-6143.2008.02444.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The use of alpha1,3-galactosyltransferase gene-knockout (GalT-KO) swine donors in discordant xenotransplantation has extended the survival of cardiac xenografts in baboons following transplantation. Eight baboons received heterotopic cardiac xenografts from GalT-KO swine and were treated with a chronic immunosuppressive regimen. The pathologic features of acute humoral xenograft rejection (AHXR), acute cellular xenograft rejection (ACXR) and chronic rejection were assessed in the grafts. No hyperacute rejection developed and one graft survived up to 6 months after transplantation. However, all GalT-KO heart grafts underwent graft failure with AHXR, ACXR and/or chronic rejection. AHXR was characterized by interstitial hemorrhage and multiple thrombi in vessels of various sizes. ACXR was characterized by TUNEL(+) graft cell injury with the infiltration of T cells (including CD3 and TIA-1(+) cytotoxic T cells), CD4(+) cells, CD8(+) cells, macrophages and a small number of B and NK cells. Chronic xenograft vasculopathy, a manifestation of chronic rejection, was characterized by arterial intimal thickening with TUNEL(+) dead cells, antibody and complement deposition, and/or cytotoxic T-cell infiltration. In conclusion, despite the absence of the Gal epitope, acute and chronic antibody and cell-mediated rejection developed in grafts, maintained by chronic immunosupression, presumably due to de novo responses to non-Gal antigens.
Collapse
Affiliation(s)
- Y. Hisashi
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - K. Yamada
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - K. Kuwaki
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Y.-L Tseng
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - F. J. M. F. Dor
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - S. L Houser
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - S. C. Robson
- Department of Medicine, Transplant Center, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA
| | | | - D. K. C. Cooper
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - D. H. Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - R. B. Colvin
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - A. Shimizu
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, Immerge BioTherapeutics Inc., Cambridge, MA, Department of Pathology, Nippon Medical School, Tokyo, Japan,Corresponding author: Akira Shimizu,
| |
Collapse
|
45
|
In Vitro Expanded Human CD4+CD25+ Regulatory T Cells are Potent Suppressors of T-Cell-Mediated Xenogeneic Responses. Transplantation 2008; 85:1841-8. [DOI: 10.1097/tp.0b013e3181734793] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Abstract
Xenotransplantation holds promise to solve the ever increasing shortage of donor organs for allotransplantation. In the last 2 decades, major progress has been made in understanding the immunobiology of pig-into-(non)human primate transplantation and today we are on the threshold of the first clinical trials. Hyperacute rejection, which is mediated by pre-existing anti-alpha Gal xenoreactive antibodies, can in non-human primates be overcome by complement- and/or antibody-modifying interventions. A major step forward was the development of genetically engineered pigs, either transgenic for human complement regulatory proteins or deficient in the alpha1,3-galactosyltranferase enzyme. However, several other immunologic and nonimmunologic hurdles remain. Acute vascular xenograft rejection is mediated by humoral and cellular mechanisms. Elicited xenoreactive antibodies play a key role. In addition to providing B cell help, xenoreactive T cells may directly contribute to xenograft rejection. Long-term survival of porcine kidney- and heart xenografts in non-human primates has been obtained but required severe T and B cell immunosuppression. Induction of xenotolerance, e.g. through mixed hematopoietic chimerism, may represent the preferred approach, but although proof of principle has been delivered in rodents, induction of pig-to-non-human primate chimerism remains problematic. Finally, it is now clear that innate immune cells, in particular macrophages and natural killer cells, can mediate xenograft destruction, the determinants of which are being elucidated. Chronic xenograft rejection is not well understood, but recent studies indicate that non-immunological problems, such as incompatibilities between human procoagulant and pig anticoagulant components may play an important role. Here, we give a comprehensive overview of the currently known obstacles to xenografting: immune and non-immune problems are discussed, as well as the possible strategies that are under development to overcome these hurdles.
Collapse
Affiliation(s)
- B Sprangers
- Laboratory of Experimental Transplantation, University of Leuven, Leuven, Belgium
| | | | | |
Collapse
|
47
|
Park CG, Kim JS, Kim YH. Current Status and Future Perspectives of Xenotransplantation and Stem Cell Research in Transplantation Field. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2008. [DOI: 10.5124/jkma.2008.51.8.732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Korea.
| | - Jung-Sik Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Korea.
| | - Yong-Hee Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Korea.
| |
Collapse
|
48
|
Yang YG, Sykes M. Xenotransplantation: current status and a perspective on the future. Nat Rev Immunol 2007; 7:519-31. [PMID: 17571072 DOI: 10.1038/nri2099] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Xenotransplantation using pigs as the transplant source has the potential to resolve the severe shortage of human organ donors. Although the development of relatively non-toxic immunosuppressive or tolerance-inducing regimens will be required to justify clinical trials using pig organs, recent advances in our understanding of the biology of xenograft rejection and zoonotic infections, and the generation of alpha1,3-galactosyltransferase-deficient pigs have moved this approach closer to clinical application. This Review highlights the major obstacles impeding the translation of xenotransplantation into clinical therapies and the potential solutions, providing a perspective on the future of clinical xenotransplantation.
Collapse
Affiliation(s)
- Yong-Guang Yang
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | |
Collapse
|
49
|
|
50
|
Abstract
Mohiuddin discusses the lessons learned from large animal xenograft models and why the immunological barrier is still the most important hurdle preventing clinical xenotransplantation of organs.
Collapse
Affiliation(s)
- Muhammad M Mohiuddin
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America.
| |
Collapse
|