1
|
Matsumura S, Kakuta Y, Maegawa-Higa Y, Fukae S, Tanaka R, Nakazawa S, Yamanaka K, Miyagawa S, Nonomura N. Differences between xenotransplantation and allogeneic kidney transplantation: the current situation and future challenges in Japan. J Artif Organs 2025:10.1007/s10047-025-01506-x. [PMID: 40316882 DOI: 10.1007/s10047-025-01506-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
Kidney transplantation is the only curative option for patients with chronic renal failure, significantly improving their survival and quality of life. However, this treatment remains limited by the shortage of organ donors. The shortage of kidney donors remains a serious problem all over the world, and is particularly severe in Japan. While advancements in immunosuppressive therapies and histocompatibility testing have improved outcomes in allogeneic kidney transplantation, the rising number of dialysis patients has worsened the gap between the demand for and supply of suitable donor organs. In response to this pressing need, xenotransplantation has gained attention as a promising alternative solution. Recent progress driven by gene-editing technologies, including CRISPR-Cas9, has facilitated the development of genetically modified pigs suitable for potential human transplantation. This review provides an overview of the key differences in immune response and infection risks between xenogeneic and allogeneic kidney transplants. In addition, it comprehensively examines the challenges and potential of xenogeneic kidney transplantation from multiple perspectives, including differences in immunosuppressive therapies between allogeneic and xenogeneic transplantation. We also discuss the feasibility of xenogeneic kidney transplantation as a solution to the organ shortage in Japan and present directions for addressing challenges toward clinical application. We hope this review will provide valuable insights into the potential of xenogeneic kidney transplantation as a new treatment option for chronic renal failure and contribute to efforts to address the donor shortage problem in Japan.
Collapse
Affiliation(s)
- Soichi Matsumura
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Yoko Maegawa-Higa
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shota Fukae
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Ryo Tanaka
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shigeaki Nakazawa
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kazuaki Yamanaka
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
2
|
Xing K, Chang Y, Zhang X, Du X, Song J. Xenotransplantation in China: Past, Present, and Future. Xenotransplantation 2025; 32:e70038. [PMID: 40243324 DOI: 10.1111/xen.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Organ failure poses a substantial global health challenge, and xenotransplantation emerges as one of the most promising avenues to mitigate the critical shortage of donor organs. In recent years, numerous research institutions have undertaken clinical and preclinical xenotransplantation in humans, instilling hope for notable progress. Nevertheless, formidable obstacles persist before success can be fully achieved. Chinese researchers have been at the forefront of xenotransplantation studies, actively contributing to several pivotal areas: the identification of critical genes essential for xenotransplantation and the creation of genetically modified pigs; preclinical studies on pig-to-nonhuman primate organ and tissue xenotransplantation, as well as the utilization of genetically engineered pig-derived biomaterials; contributions to both preclinical and clinical xenotransplantation research; and the formulation and refinement of xenotransplantation policies and ethical guidelines in China. In conclusion, this review seeks to not only acknowledge the contributions of Chinese researchers but also to encourage further collaboration between Chinese scholars and their international counterparts in advancing the field of xenotransplantation.
Collapse
Affiliation(s)
- Kai Xing
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Yuan Chang
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiulin Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingchao Du
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Jiangping Song
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| |
Collapse
|
3
|
Nascimento H, Martins TMM, Moreira R, Barbieri G, Pires P, Carvalho LN, Rosa LR, Almeida A, Araujo MS, Pessuti CL, Ferrer H, Pereira Gomes JÁ, Belfort R, Raia S. Current Scenario and Future Perspectives of Porcine Corneal Xenotransplantation. Cornea 2025; 44:387-404. [PMID: 39413247 DOI: 10.1097/ico.0000000000003723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/25/2024] [Indexed: 10/18/2024]
Abstract
ABSTRACT Corneal diseases represent a significant cause of blindness worldwide, with corneal transplantation being an effective treatment to prevent vision loss. Despite substantial advances in transplantation techniques, the demand for donor corneas exceeds the available supply, particularly in developing countries. Cornea xenotransplantation has emerged as a promising strategy to address the worldwide scarcity, notably using porcine corneas. In addition to the inherent immune privilege of the cornea, the low cost of porcine breeding and the anatomical and physiological similarities between humans and pigs have made porcine corneas a viable alternative. Nonetheless, ethical concerns, specifically the risk of xenozoonotic transmission and the necessity for stringent biosafety measures, remain significant obstacles. Moreover, the success of xenotransplantation is compromised by innate and adaptive immune responses, which requires meticulous consideration and further studies. Despite these challenges, recent breakthroughs have further contributed to reducing immunogenicity while preserving the corneal architecture. Advances in genetic engineering, such as the use of CRISPR-Cas9 to eliminate critical porcine antigens, have shown promise for mitigating immune reactions. Additionally, new immunosuppressive protocols, such as have techniques like decellularization and the use of porcine-derived acellular matrices, have greatly increased graft survival in preclinical models. Future research must focus on refining immunomodulatory strategies and improving graft preparation techniques to ensure the long-term survival and safety of porcine corneal xenotransplantation in clinical trials in humans.
Collapse
Affiliation(s)
- Heloisa Nascimento
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
| | - Thaís M M Martins
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
- Federal University of Viçosa (UFV), Viçosa, Brazil; and
| | | | - Gabriel Barbieri
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Pedro Pires
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
| | - Lucimeire N Carvalho
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Larissa R Rosa
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Augusto Almeida
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
| | | | - Carmen Luz Pessuti
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
| | - Henrique Ferrer
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
| | | | - Rubens Belfort
- Department of Ophthalmology, Federal University of São Paulo (UNIFESP), Sao Paulo, Brazil
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
- Federal University of Viçosa (UFV), Viçosa, Brazil; and
- Vision Institute (IPEPO), Sao Paulo, Brazil
| | - Silvano Raia
- Faculty of Medicine, University of São Paulo (USP), Sao Paulo, Brazil
| |
Collapse
|
4
|
Deng S, Zhang Y, Shen S, Li C, Qin C. Immunometabolism of Liver Xenotransplantation and Prospective Solutions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407610. [PMID: 39912334 PMCID: PMC11884532 DOI: 10.1002/advs.202407610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/26/2024] [Indexed: 02/07/2025]
Abstract
End-stage liver diseases, such as hepatocellular carcinoma or acute liver failure, critically necessitate liver transplantation. However, the shortage of available organ donors fails to meet the rapidly growing transplantation demand. Due to the high similarity of liver tissue structure and metabolism between miniature pigs and humans, xenotransplantation of pig livers is considered as a potentially viable solution to organ scarcity. In the 2024, teams from China first time have successfully transplanted a genetically modified Bama miniature pig liver into a clinically brain-dead man lasting for 10 days. This milestone in human xenotransplantation research not only confirms the feasibility of clinical application of xenotransplantation, but also underscores the daunting and protracted nature of this pathway. Despite advanced gene-editing technologies theoretically circumventing the occurrence of most transplant rejection reactions, patients still face challenges such as chronic immune rejection, coagulation disorders, and thrombotic microangiopathy after receiving xenografts. Moreover, prolonged use of immunosuppressive drugs may induce irreversible immune dysfunction, leading to opportunistic infections and metabolic disorders. This article compares the similarities and differences in livers between humans and pigs, summarizes the immunometabolism of xenotransplantation based on current findings, and provides research perspectives on pre-transplantation and post-transplantation strategies for prolonging the survival time of xenografts.
Collapse
Affiliation(s)
- Shoulong Deng
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijing100021China
| | - Yi Zhang
- Department of MedicinePanzhihua UniversitySichuan61700China
| | - Shasha Shen
- Department of MedicinePanzhihua UniversitySichuan61700China
| | - Chongyang Li
- Institute of Animal SciencesChinese Academy of Agricultural SciencesBeijing100193China
| | - Chuan Qin
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijing100021China
| |
Collapse
|
5
|
Galdina V, Puga Yung GL, Seebach JD. Cytotoxic Responses Mediated by NK Cells and Cytotoxic T Lymphocytes in Xenotransplantation. Transpl Int 2025; 38:13867. [PMID: 40012743 PMCID: PMC11862997 DOI: 10.3389/ti.2025.13867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/09/2025] [Indexed: 02/28/2025]
Abstract
Xenotransplantation represents a potential solution to the shortage of organs for transplantation. The recent advancements in porcine genetic modification have addressed hyperacute and acute vascular rejection; however, challenges persist with regard to delayed xenograft rejection. Porcine endothelial cells (pECs) represent a crucial target in the context of xenograft rejection, which is mediated by cytotoxic lymphocytes. It is crucial to comprehend the manner in which human natural killer (NK) cells and cytotoxic CD8+ T lymphocytes (CTL) recognize and target pECs in order to develop efficacious prophylactic strategies against rejection. The objective of the present review is to synthesize the existing knowledge regarding the mechanisms and techniques employed to modulate xenogeneic responses mediated by human NK cells and CTL. We will elucidate recent methodological advancements, debate potential novel strategies, and emphasize the imperative necessity for further research and innovative approaches to enhance graft survival.
Collapse
|
6
|
Kale A, Rogers NM. Research Highlights. Transplantation 2025; 109:98-101. [PMID: 39700178 DOI: 10.1097/tp.0000000000005285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Affiliation(s)
- Atharva Kale
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Health and Medicine, University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Health and Medicine, University of Sydney, Camperdown, Sydney, NSW, Australia
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
7
|
Kakuta Y, Miyagawa S, Matsumura S, Higa-Maegawa Y, Fukae S, Tanaka R, Nakazawa S, Yamanaka K, Kawamura T, Saito S, Miyagawa S, Nonomura N. Complement and complement regulatory protein in allogeneic and xenogeneic kidney transplantation. Transplant Rev (Orlando) 2025; 39:100885. [PMID: 39536474 DOI: 10.1016/j.trre.2024.100885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Kidney transplantation is the most optimal treatment for patients with end-stage renal disease, offering significant improvements in patient outcomes over dialysis. However, the potential for immune rejection, where the recipient's immune system attacks the transplanted kidney, can compromise transplant success. The complement system, a key component of the immune response, plays a crucial role in both acute and chronic rejection, including T-cell- and antibody-mediated rejection. Understanding and controlling the complement system is essential for managing rejection and enhancing graft survival and overall success of kidney transplantation. In allogeneic transplantation, complement activation through various pathways contributes to graft damage and failure. Recent advancements in genetic engineering enable the development of transgenic pigs expressing human complement regulatory proteins, which display potential for reducing rejection in xenotransplantation. Despite these advances, the complex mechanisms of complement activation and regulation are not fully understood, necessitating further research. This review examines the role of the complement system in kidney transplantation, explores the latest developments in complement regulatory strategies, and discusses potential therapeutic approaches to improve transplant outcomes.
Collapse
Affiliation(s)
- Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Japan.
| | - Soichi Matsumura
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Yoko Higa-Maegawa
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Shota Fukae
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Ryo Tanaka
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Shigeaki Nakazawa
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Kazuaki Yamanaka
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Japan
| | - Shunsuke Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
8
|
Fang B, Wang C, Yuan Y, Liu X, Shi L, Li L, Wang Y, Dai Y, Yang H. Generation and characterization of genetically modified pigs with GGTA1/β4GalNT2/CMAH knockout and human CD55/CD47 expression for xenotransfusion studies. Sci Rep 2024; 14:29870. [PMID: 39622959 PMCID: PMC11612173 DOI: 10.1038/s41598-024-81730-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Pig red blood cells (pRBCs) represent a promising alternative to address the shortage in transfusion medicine. Nonetheless, a major obstacle to their clinical implementation is immunological rejection. In this study, we generated transgenic pigs expressing human CD47 (hCD47) and CD55 (hCD55) in α1,3-galactosyltransferase KO/β-1,4-N-acetyl-galactosaminyl transferase 2 KO/cytidine monophosphate-N-acetylneuraminic acid hydroxylase KO (TKO) pigs using CRISPR/Cas9 technology. Compared to wild-type pRBCs, TKO/hCD47/hCD55 pRBCs exhibit significantly reduced human IgG/IgM antibody binding. Moreover, when transfused into Cynomolgus monkeys, TKO/hCD47/hCD55 pRBCs remained detectable for 2 h post-transfusion, whereas wild-type pRBCs were eliminated within 20 min. This study demonstrates the potential of multi-gene edited pigs to provide immunologically compatible pRBCs.
Collapse
Affiliation(s)
- Bin Fang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
- Gcreatene (Suzhou) Biotechnology Co., Ltd., Suzhou, 215000, China
| | - Chunting Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Yilin Yuan
- Gcreatene (Suzhou) Biotechnology Co., Ltd., Suzhou, 215000, China
| | - Xiaorui Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China
| | - Lili Shi
- Jiangsu Province Blood Center, Nanjing, 210042, China
| | - Lin Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China.
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China.
- Gcreatene (Suzhou) Biotechnology Co., Ltd., Suzhou, 215000, China.
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Haiyuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China.
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
9
|
Wang W, Liu Z, Zhu J, Zhen H, Qi M, Luo J, Zhen J. Macrophage tracking with USPIO imaging and T2 mapping predicts immune rejection of transplanted stem cells. Sci Rep 2024; 14:29162. [PMID: 39587241 PMCID: PMC11589617 DOI: 10.1038/s41598-024-80750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
To develop a clinical imaging method for monitoring macrophage migration to the defect site after implantation of various stem cells and evaluating immune responses in the context of knee arthritis, T2 mapping was correlated with CD68-positive cell densities in defects and the bone marrow. This study, which was approved by the Institutional Animal Care and Use Committee, used 32 New Zealand white rabbits preloaded with ultrasmall superparamagnetic iron oxide particles (USPIOs). They were divided into groups that received different stem cell implants after osteochondral defect induction. T2 imaging was performed using a 3.0 T MR scanner, and the data were analysed via one-way ANOVA, with CD68 expression assessed via immunohistochemistry. After implantation, the T2 signal intensity increased across groups, with subgroup D1 (implantation of rat bone marrow stem cells (BMSCs)) showing the lowest T2 value early and the steepest increase in T2 values. Notable differences in CD68-positive cell density were found between Subgroup D1 and the other groups and between Subgroups A1 and C1 post-surgery. A moderate negative correlation was observed between T2 signals and CD68-positive cell density in defects (r = -0.468, p = 0.001), whereas a weak correlation was detected in the bone marrow (r = 0.096, p = 0.313). A significant link was identified between CD68-positive cell density in the bone marrow and in defects (r = -0.255, p = 0.001). This study revealed significant differences in immune responses to stem cells from different origin tissues in the context of cartilage repair. Adipose-derived stem cells (ADSCs) were found to be more likely to provoke immune rejection than were BMSCs in the repair of femoral condyle cartilage defects. Compared with allogeneic transplants, xenogeneic mesenchymal stem cell transplants were associated with prolonged immune rejection. T2 mapping technology was effective in predicting the density of CD68-positive cells, providing a valuable tool for immune monitoring in stem cell therapy.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, 116000, Liaoning, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhenyu Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jiahong Zhu
- Department of Radiology, Taiyuan Hospital of Traditional Chinese Medicine, Taiyuan, 030001, Shanxi, China
| | - Haocheng Zhen
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Meiling Qi
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jing Luo
- Department of Rheumatology and immunology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junping Zhen
- Department of Rheumatology and immunology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Department of Magnetic Resonance, Faculty of Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
10
|
Sakano Y, Sakano K, Hurrell BP, Shafiei-Jahani P, Kazemi MH, Li X, Shen S, Barbers R, Akbari O. SIRPα engagement regulates ILC2 effector function and alleviates airway hyperreactivity via modulating energy metabolism. Cell Mol Immunol 2024; 21:1158-1174. [PMID: 39160226 PMCID: PMC11442993 DOI: 10.1038/s41423-024-01208-z] [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: 06/02/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Group-2 innate lymphoid cells (ILC2) are part of a growing family of innate lymphocytes known for their crucial role in both the development and exacerbation of allergic asthma. The activation and function of ILC2s are regulated by various activating and inhibitory molecules, with their balance determining the severity of allergic responses. In this study, we aim to elucidate the critical role of the suppressor molecule signal regulatory protein alpha (SIRPα), which interacts with CD47, in controlling ILC2-mediated airway hyperreactivity (AHR). Our data indicate that activated ILC2s upregulate the expression of SIRPα, and the interaction between SIRPα and CD47 effectively suppresses both ILC2 proliferation and effector function. To evaluate the function of SIRPα in ILC2-mediated AHR, we combined multiple approaches including genetically modified mouse models and adoptive transfer experiments in murine models of allergen-induced AHR. Our findings suggest that the absence of SIRPα leads to the overactivation of ILC2s. Conversely, engagement of SIRPα with CD47 reduces ILC2 cytokine production and effectively regulates ILC2-dependent AHR. Furthermore, the SIRPα-CD47 axis modulates mitochondrial metabolism through the JAK/STAT and ERK/MAPK signaling pathways, thereby regulating NF-κB activity and the production of type 2 cytokines. Additionally, our studies have revealed that SIRPα is inducible and expressed on human ILC2s, and administration of human CD47-Fc effectively suppresses the effector function and cytokine production. Moreover, administering human CD47-Fc to humanized ILC2 mice effectively alleviates AHR and lung inflammation. These findings highlight the promising therapeutic potential of targeting the SIRPα-CD47 axis in the treatment of ILC2-dependent allergic asthma.
Collapse
Affiliation(s)
- Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mohammad Hossein Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Shen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Richard Barbers
- Department of Clinical Medicine, Division of Pulmonary and Critical Care Medicine, Keck School of Medicine of USC, University of Southern California Hospital, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Saito S, Miyagawa S, Kawamura T, Yoshioka D, Kawamura M, Kawamura A, Misumi Y, Taguchi T, Yamauchi T, Miyagawa S. How should cardiac xenotransplantation be initiated in Japan? Surg Today 2024; 54:829-838. [PMID: 38733536 PMCID: PMC11266268 DOI: 10.1007/s00595-024-02861-7] [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: 02/13/2024] [Accepted: 04/07/2024] [Indexed: 05/13/2024]
Abstract
The world's first clinical cardiac xenotransplantation, using a genetically engineered pig heart with 10 gene modifications, prolonged the life of a 57-year-old man with no other life-saving options, by 60 days. It is foreseeable that xenotransplantation will be introduced in clinical practice in the United States. However, little clinical or regulatory progress has been made in the field of xenotransplantation in Japan in recent years. Japan seems to be heading toward a "device lag", and the over-importation of medical devices and technology in the medical field is becoming problematic. In this review, we discuss the concept of pig-heart xenotransplantation, including the pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental heart overgrowth, as well as genetic modification strategies in pigs to prevent or minimize these problems. Moreover, we summarize the necessity for and current status of xenotransplantation worldwide, and future prospects in Japan, with the aim of initiating xenotransplantation in Japan using genetically modified pigs without a global delay. It is imperative that this study prompts the initiation of preclinical xenotransplantation research using non-human primates and leads to clinical studies.
Collapse
Affiliation(s)
- Shunsuke Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Yoshioka
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masashi Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Misumi
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | - Takashi Yamauchi
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
12
|
Grimus S, Sarangova V, Welzel PB, Ludwig B, Seissler J, Kemter E, Wolf E, Ali A. Immunoprotection Strategies in β-Cell Replacement Therapy: A Closer Look at Porcine Islet Xenotransplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401385. [PMID: 38884159 PMCID: PMC11336975 DOI: 10.1002/advs.202401385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/28/2024] [Indexed: 06/18/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by absolute insulin deficiency primarily due to autoimmune destruction of pancreatic β-cells. The prevailing treatment for T1DM involves daily subcutaneous insulin injections, but a substantial proportion of patients face challenges such as severe hypoglycemic episodes and poorly controlled hyperglycemia. For T1DM patients, a more effective therapeutic option involves the replacement of β-cells through allogeneic transplantation of either the entire pancreas or isolated pancreatic islets. Unfortunately, the scarcity of transplantable human organs has led to a growing list of patients waiting for an islet transplant. One potential alternative is xenotransplantation of porcine pancreatic islets. However, due to inter-species molecular incompatibilities, porcine tissues trigger a robust immune response in humans, leading to xenograft rejection. Several promising strategies aim to overcome this challenge and enhance the long-term survival and functionality of xenogeneic islet grafts. These strategies include the use of islets derived from genetically modified pigs, immunoisolation of islets by encapsulation in biocompatible materials, and the creation of an immunomodulatory microenvironment by co-transplanting islets with accessory cells or utilizing immunomodulatory biomaterials. This review concentrates on delineating the primary obstacles in islet xenotransplantation and elucidates the fundamental principles and recent breakthroughs aimed at addressing these challenges.
Collapse
Affiliation(s)
- Sarah Grimus
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| | - Victoria Sarangova
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Petra B. Welzel
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Barbara Ludwig
- Department of Medicine IIIUniversity Hospital Carl Gustav CarusTechnische Universität DresdenD‐01307DresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the Technische Universität DresdenD‐01307DresdenGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
- DFG‐Center for Regenerative Therapies DresdenTechnische Universität DresdenD‐01307DresdenGermany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IVDiabetes Zentrum – Campus InnenstadtKlinikum der Ludwig‐Maximilians‐Universität MünchenD‐80336MunichGermany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Asghar Ali
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| |
Collapse
|
13
|
Peterson L, Yacoub MH, Ayares D, Yamada K, Eisenson D, Griffith BP, Mohiuddin MM, Eyestone W, Venter JC, Smolenski RT, Rothblatt M. Physiological basis for xenotransplantation from genetically modified pigs to humans. Physiol Rev 2024; 104:1409-1459. [PMID: 38517040 PMCID: PMC11390123 DOI: 10.1152/physrev.00041.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
The collective efforts of scientists over multiple decades have led to advancements in molecular and cellular biology-based technologies including genetic engineering and animal cloning that are now being harnessed to enhance the suitability of pig organs for xenotransplantation into humans. Using organs sourced from pigs with multiple gene deletions and human transgene insertions, investigators have overcome formidable immunological and physiological barriers in pig-to-nonhuman primate (NHP) xenotransplantation and achieved prolonged pig xenograft survival. These studies informed the design of Revivicor's (Revivicor Inc, Blacksburg, VA) genetically engineered pigs with 10 genetic modifications (10 GE) (including the inactivation of 4 endogenous porcine genes and insertion of 6 human transgenes), whose hearts and kidneys have now been studied in preclinical human xenotransplantation models with brain-dead recipients. Additionally, the first two clinical cases of pig-to-human heart xenotransplantation were recently performed with hearts from this 10 GE pig at the University of Maryland. Although this review focuses on xenotransplantation of hearts and kidneys, multiple organs, tissues, and cell types from genetically engineered pigs will provide much-needed therapeutic interventions in the future.
Collapse
Affiliation(s)
- Leigh Peterson
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | | | - David Ayares
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - Kazuhiko Yamada
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Daniel Eisenson
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Bartley P Griffith
- University of Maryland Medical Center, Baltimore, Maryland, United States
| | | | - Willard Eyestone
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - J Craig Venter
- J. Craig Venter Institute, Rockville, Maryland, United States
| | | | - Martine Rothblatt
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| |
Collapse
|
14
|
Iemitsu K, Sakai R, Maeda A, Gadomska K, Kogata S, Yasufuku D, Matsui J, Masahata K, Kamiyama M, Eguchi H, Matsumura S, Kakuta Y, Nagashima H, Okuyama H, Miyagawa S. The hybrid CL-SP-D molecule has the potential to regulate xenogeneic rejection by human neutrophils more efficiently than CD47. Transpl Immunol 2024; 84:102020. [PMID: 38452982 DOI: 10.1016/j.trim.2024.102020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Innate immunity plays a vital role in xenotransplantation. A CD47 molecule, binding to the SIRPα expressed on monocyte/macrophage cells, can suppress cytotoxicity. Particularly, the SIRPα contains ITIM, which delivers a negative signal. Our previous study demonstrated that the binding between CL-P1 and surfactant protein-D hybrid (CL-SP-D) with SIRPα regulates macrophages' phagocytic activity. In this study, we examined the effects of human CD47 and CL-SP-D expression on the inhibition of xenograft rejection by neutrophils in swine endothelial cells (SECs). METHODS We first examined SIRPα expression on HL-60 cells, a neutrophil-like cell line, and neutrophils isolated from peripheral blood. CD47-expressing SECs or CL-SP-D-expressing SECs were generated through plasmid transfection. Subsequently, these SECs were co-cultured with HL-60 cells or neutrophils. After co-culture, the degree of cytotoxicity was calculated using the WST-8 assay. The suppressive function of CL-SP-D on neutrophils was subsequently examined, and the results were compared with those of CD47 using naïve SECs as controls. Additionally, we assessed ROS production and neutrophil NETosis. RESULTS In initial experiments, the expression of SIRPα on HL-60 and neutrophils was confirmed. Exposure to CL-SP-D significantly suppressed the cytotoxicity in HL-60 (p = 0.0038) and neutrophils (p = 0.00003). Furthermore, engagement with CD47 showed a suppressive effect on neutrophils obtained from peripheral blood (p = 0.0236) but not on HL-60 (p = 0.4244). The results of the ROS assays also indicated a significant downregulation of SEC by CD47 (p = 0.0077) or CL-SP-D (p = 0.0018). Additionally, the suppression of NETosis was confirmed (p = 0.0125) in neutrophils co-cultured with S/CL-SP-D. CONCLUSION These results indicate that CL-SP-D is highly effective on neutrophils in xenogeneic rejection. Furthermore, CL-SP-D was more effective than CD47 at inhibiting neutrophil-mediated xenograft rejection.
Collapse
Affiliation(s)
- Keigo Iemitsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa, Japan
| | - Rieko Sakai
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa, Japan
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Katarzyna Gadomska
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Daiki Yasufuku
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jun Matsui
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Soichi Matsumura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa, Japan.
| |
Collapse
|
15
|
Vadori M, Cozzi E. Current challenges in xenotransplantation. Curr Opin Organ Transplant 2024; 29:205-211. [PMID: 38529696 PMCID: PMC11064916 DOI: 10.1097/mot.0000000000001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
PURPOSE OF REVIEW In recent years, the xenotransplantation science has advanced tremendously, with significant strides in both preclinical and clinical research. This review intends to describe the latest cutting-edge progress in knowledge and methodologies developed to overcome potential obstacles that may preclude the translation and successful application of clinical xenotransplantation. RECENT FINDINGS Preclinical studies have demonstrated that it is now possible to extend beyond two years survival of primate recipients of life saving xenografts. This has been accomplished thanks to the utilization of genetic engineering methodologies that have allowed the generation of specifically designed gene-edited pigs, a careful donor and recipient selection, and appropriate immunosuppressive strategies.In this light, the compassionate use of genetically modified pig hearts has been authorized in two human recipients and xenotransplants have also been achieved in human decedents. Although encouraging the preliminary results suggest that several challenges have yet to be fully addressed for a successful clinical translation of xenotransplantation. These challenges include immunologic, physiologic and biosafety aspects. SUMMARY Recent progress has paved the way for the initial compassionate use of pig organs in humans and sets the scene for a wider application of clinical xenotransplantation.
Collapse
Affiliation(s)
- Marta Vadori
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua
| | - Emanuele Cozzi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua
- Transplant Immunology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health Padua University Hospital, Padua, Italy
| |
Collapse
|
16
|
Kim B, Yan JJ, Kang TK, Lee WB, Jeong JC, Yang J. Molecular incompatibility between pig CD200 and human CD200 receptor in in vitro xenogeneic immune responses. Xenotransplantation 2024; 31:e12863. [PMID: 38751087 DOI: 10.1111/xen.12863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/22/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Overexpression of human CD200 (hCD200) in porcine endothelial cells (PECs) has been reported to suppress xenogeneic immune responses of human macrophages against porcine endothelial cells. The current study aimed to address whether the above-mentioned beneficial effect of hCD200 is mediated by overcoming the molecular incompatibility between porcine CD200 (pCD200) and hCD200 receptor or simply by increasing the expression levels of CD200 without any molecular incompatibility across the two species. We overexpressed hCD200 or pCD200 using lentiviral vectors with V5 marker in porcine endothelial cells and compared their suppressive activity against U937-derived human macrophage-like cells (hMCs) and primary macrophages. In xenogeneic coculture of porcine endothelial cells and human macrophage-like cells or macrophages, hCD200-porcine endothelial cells suppressed phagocytosis and cytotoxicity of human macrophages to a greater extent than pCD200-porcine endothelial cells. Secretion of tumor necrosis factor-α, interleukin-1β, and monocyte chemoattractant protein-1 from human macrophages and expression of M1 phenotypes (inducible nitric oxide synthase, dectin-1, and CD86) were also suppressed by hCD200 to a greater extent than pCD200. Furthermore, in signal transduction downstream of CD200 receptor, hCD200 induced Dok2 phosphorylation and suppressed IκB phosphorylation to a greater extent than pCD200. The above data supported the possibility of a significant molecular incompatibility between pCD200 and human CD200 receptor, suggesting that the beneficial effects of hCD200 overexpression in porcine endothelial cells could be mediated by overcoming the molecular incompatibility across the species barrier rather than by simple overexpression effects of CD200.
Collapse
Affiliation(s)
- Bomin Kim
- Graduate School of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Jing Yan
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae Kyeom Kang
- Natural Product Research Center, Korea Institute of Science & Technology, Gangneung, Republic of Korea
| | - Wook-Bin Lee
- Natural Product Research Center, Korea Institute of Science & Technology, Gangneung, Republic of Korea
| | - Jong Cheol Jeong
- Department of Internal Medicine, Seoul National University College of Medicine, Bundang Hospital, Seoul, Republic of Korea
| | - Jaeseok Yang
- Graduate School of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Zhao C, Wang C, Shan W, Wang Z, Chen X, Deng H. Nanomedicines for an Enhanced Immunogenic Cell Death-Based In Situ Cancer Vaccination Response. Acc Chem Res 2024; 57:905-918. [PMID: 38417027 DOI: 10.1021/acs.accounts.3c00771] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Cancer vaccines have shown tremendous potential in preventing and treating cancer by providing immunogenic antigens to initiate specific tumor immune responses. An in situ vaccine prepared from an autologous tumor can mobilize a patient's own tumor cell lysate as a reservoir of specific antigens, thus triggering a broad immune response and diverse antitumor immunity in an individually tailored manner. Its efficacy is much better than that of conventional vaccines with a limited number of epitopes. Several conventional therapies, including radiotherapy (RT), chemotherapeutics, photodynamic therapy (PDT), and photothermal therapy (PTT) can activate an anticancer in situ vaccine response by inducing immunogenic cell death (ICD), triggering the exposure of tumor-associated antigens (TAAs), cancerous testis antigens, neoantigens, and danger-associated molecular patterns (DAMPs) with low cost. However, the immunogenicity of dying tumor cells is low, making released antigens and DAMPs insufficient to initiate a robust immune response against malignant cancer. Moreover, the immunosuppressive tumor microenvironment (TME) severely hinders the infiltration and sensitization of effector immune cells, causing tolerogenic immunological effects.Herein, we mainly focus on the research in developing nanoplatforms to surmount the major challenges met by ICD-based in situ vaccines. We first summarized a variety of nanotechnologies that enable enhanced immunogenicity of dying cancer cells by enhancing antigenicity and adjuvanticity. The robust antigenicity was obtained via regulating the tumor cells death mode or the dying state to amplify the recognition of tumor debris by professional antigen-presenting cells (APCs). The adjuvanticity was potentiated by raising the level or intensifying the activity of endogenous adjuvants or promoting the intelligent delivery of exogenous immunostimulants to activate immune cell recruitment and promote antigen presentation. Additionally, versatile approaches to reverse immunosuppressive TME to boost the in situ tumor vaccination response are also highlighted in detail. On one hand, by modulating the cell metabolism in TME, the expansion and activity of effector versus immunosuppressive cells can be optimized to improve the efficiency of in situ vaccines. On the other hand, regulating cellular components in TME, such as reversing adverse immune cell phenotypes or inhibiting the activity of interstitial cells, can also significantly enhance the ICD-based antitumor immunotherapy effect. Finally, our viewpoint on the future challenges and opportunities in this hopeful area is presented. We expect that this Account can offer much more insight into the design, planning, and development of cutting-edge in situ tumor vaccine platforms, promoting more attention and academic-industry collaborations, accelerating the advanced progress of in situ tumor vaccine-based immunotherapy in the clinic.
Collapse
Affiliation(s)
- Caiyan Zhao
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Changrong Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Wenbo Shan
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhongliang Wang
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Hongzhang Deng
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of Trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
18
|
Xu L, Wang X, Zhang T, Meng X, Zhao W, Pi C, Yang YG. Expression of a mutant CD47 protects against phagocytosis without inducing cell death or inhibiting angiogenesis. Cell Rep Med 2024; 5:101450. [PMID: 38508139 PMCID: PMC10983038 DOI: 10.1016/j.xcrm.2024.101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/22/2023] [Accepted: 02/09/2024] [Indexed: 03/22/2024]
Abstract
CD47 is a ligand of SIRPα, an inhibitory receptor expressed by macrophages, dendritic cells, and natural killer (NK) cells, and, therefore, transgenic overexpression of CD47 is considered an effective approach to inhibiting transplant rejection. However, the detrimental effect of CD47 signaling is overlooked when exploring this approach. Here, we construct a mutant CD47 by replacing the transmembrane and intracellular domains with a membrane anchor (CD47-IgV). In both human and mouse cells, CD47-IgV is efficiently expressed on the cell surface and protects against phagocytosis in vitro and in vivo but does not induce cell death or inhibit angiogenesis. Furthermore, hematopoietic stem cells expressing transgenic CD47-IgV show no detectable alterations in engraftment or differentiation. This study provides a potentially effective means of achieving transgenic CD47 expression that may help to produce gene-edited pigs for xenotransplantation and hypoimmunogenic pluripotent stem cells for regenerative medicine.
Collapse
Affiliation(s)
- Lu Xu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Xiaodan Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Ting Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Wenjie Zhao
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Chenchen Pi
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, First Hospital of Jilin University, Changchun, Jilin 130062, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, Jilin University, Changchun, Jilin 130062, China; International Center of Future Science, Jilin University, Changchun, Jilin 130062, China.
| |
Collapse
|
19
|
Paldino E, Migliorato G, Fusco FR. Neuroimmune pathways involvement in neurodegeneration of R6/2 mouse model of Huntington's disease. Front Cell Neurosci 2024; 18:1360066. [PMID: 38444595 PMCID: PMC10912295 DOI: 10.3389/fncel.2024.1360066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024] Open
Abstract
Mechanisms of tissue damage in Huntington's disease (HD) involve excitotoxicity, mitochondrial damage, and neuroinflammation, including microglia activation. CD47 is a membrane protein that interacts with the inhibitory immunoreceptor SIRPα. Engagement of SIRPα by CD47 provides a downregulatory signal that inhibits host cell phagocytosis, promoting a "don't-eat-me" signal. These proteins are involved in the immune response and are downmodulated in inflammatory diseases. The involvement of inflammation and of the inflammasome in HD has already been described. In this study, we focused on other factors that can be involved in the unregulated inflammatory response that accelerates and exacerbate the neurodegenerative process in HD. Our results show that CD47 on striatal neurons decreased in HD mice, while it increased in wild type mice with age. SIRPα, on the other hand, was present in neurons in the wild type and increases in the R6/2 mice at all stages. Recruitment of SIRPα and binding to CD47 promotes the activation through phosphorylating events of non-receptor protein tyrosine phosphatase SHP-1 and SHP-2 in neurons and microglia. SHP phosphatases are able to curb the activity of NLRP3 inflammasome thereby reducing the detrimental effect of neuroinflammation. Such activity is mediated by the inhibition (dephosphorylation) of the proteins signal transducer and activator of transcription (STAT). We found that activated SHP-1 was present in microglia and neurons of WT mice at 5 and 13 weeks, increasing with time; while in R6/2 it was not localized in neurons but only in microglia, where it decreases with time. Consequently, STAT1 was overexpressed in neurons of R6/2 mice, as an effect of lack of modulation by SHP-1. Thus, our results shed light on the pathophysiology of neuronal damage, on one hand, paving the way toward a modulation of signal transducer proteins by specific inhibitors to achieve neuroprotection in HD, on the other.
Collapse
Affiliation(s)
- Emanuela Paldino
- Laboratory of Neuroanatomy, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Giorgia Migliorato
- Laboratory of Neuroanatomy, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | |
Collapse
|
20
|
Isenberg JS, Montero E. Tolerating CD47. Clin Transl Med 2024; 14:e1584. [PMID: 38362603 PMCID: PMC10870051 DOI: 10.1002/ctm2.1584] [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: 11/19/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Cluster of differentiation 47 (CD47) occupies the outer membrane of human cells, where it binds to soluble and cell surface receptors on the same and other cells, sculpting their topography and resulting in a pleiotropic receptor-multiligand interaction network. It is a focus of drug development to temper and accentuate CD47-driven immune cell liaisons, although consideration of on-target CD47 effects remain neglected. And yet, a late clinical trial of a CD47-blocking antibody was discontinued, existent trials were restrained, and development of CD47-targeting agents halted by some pharmaceutical companies. At this point, if CD47 can be exploited for clinical advantage remains to be determined. Herein an airing is made of the seemingly conflicting actions of CD47 that reflect its position as a junction connecting receptors and signalling pathways that impact numerous human cell types. Prospects of CD47 boosting and blocking are considered along with potential therapeutic implications for autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Jeffrey S. Isenberg
- Department of Diabetes Complications & MetabolismArthur Riggs Diabetes & Metabolism Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Enrique Montero
- Department of Molecular & Cellular EndocrinologyArthur Riggs Diabetes & Metabolism Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| |
Collapse
|
21
|
Du EJ, Muench MO. A Monocytic Barrier to the Humanization of Immunodeficient Mice. Curr Stem Cell Res Ther 2024; 19:959-980. [PMID: 37859310 PMCID: PMC10997744 DOI: 10.2174/011574888x263597231001164351] [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: 06/25/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Mice with severe immunodeficiencies have become very important tools for studying foreign cells in an in vivo environment. Xenotransplants can be used to model cells from many species, although most often, mice are humanized through the transplantation of human cells or tissues to meet the needs of medical research. The development of immunodeficient mice is reviewed leading up to the current state-of-the-art strains, such as the NOD-scid-gamma (NSG) mouse. NSG mice are excellent hosts for human hematopoietic stem cell transplants or immune reconstitution through transfusion of human peripheral blood mononuclear cells. However, barriers to full hematopoietic engraftment still remain; notably, the survival of human cells in the circulation is brief, which limits overall hematological and immune reconstitution. Reports have indicated a critical role for monocytic cells - monocytes, macrophages, and dendritic cells - in the clearance of xenogeneic cells from circulation. Various aspects of the NOD genetic background that affect monocytic cell growth, maturation, and function that are favorable to human cell transplantation are discussed. Important receptors, such as SIRPα, that form a part of the innate immune system and enable the recognition and phagocytosis of foreign cells by monocytic cells are reviewed. The development of humanized mouse models has taken decades of work in creating more immunodeficient mice, genetic modification of these mice to express human genes, and refinement of transplant techniques to optimize engraftment. Future advances may focus on the monocytic cells of the host to find ways for further engraftment and survival of xenogeneic cells.
Collapse
Affiliation(s)
- Emily J. Du
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
| | - Marcus O. Muench
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
22
|
Feng Y, Huang C, Wang Y, Chen J. SIRPα: A key player in innate immunity. Eur J Immunol 2023; 53:e2350375. [PMID: 37672390 DOI: 10.1002/eji.202350375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/15/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
Signal regulatory protein alpha (SIRPα) is a crucial inhibitory regulator expressed on the surface of myeloid cells, including macrophages, dendritic cells, monocytes, neutrophils, and microglia. SIRPα plays an indispensable role in innate immune and adoptive immune responses in cancer immunology, tissue homeostasis, and other physiological or phycological conditions. This review provides an overview of the research history, ligands, signal transduction pathways, and functional mechanisms associated with SIRPα. Additionally, we summarize the therapeutic implications of targeting SIRPα as a promising novel strategy in immuno-oncology.
Collapse
Affiliation(s)
- Yongyi Feng
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunliu Huang
- Molecular Imaging Center, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yingzhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
23
|
Singireddy S, Tully A, Galindo J, Ayares D, Singh AK, Mohiuddin MM. Genetic Engineering of Donor Pig for the First Human Cardiac Xenotransplantation: Combatting Rejection, Coagulopathy, Inflammation, and Excessive Growth. Curr Cardiol Rep 2023; 25:1649-1656. [PMID: 37938425 DOI: 10.1007/s11886-023-01978-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/09/2023]
Abstract
PURPOSE OF REVIEW The first successful pig to human cardiac xenotransplantation in January 2022 represented a major step forward in the fields of heart failure, immunology, and applied genetic engineering, using a 10-gene edited (GE) pig. This review summarizes the evolution of preclinical modelling data which informed the use of each of the 10 genes modified in the 10-GE pig: GGTA1, Β4GalNT2, CMAH, CD46, CD55, TBM, EPCR, CD47, HO-1, and growth hormone receptor. RECENT FINDINGS The translation of the 10-GE pig from preclinical modelling to clinical compassionate xenotransplant use was the culmination of decades of research combating rejection, coagulopathy, inflammation, and excessive xenograft growth. Understanding these 10 genes with a view to their combinatorial effects will be useful in anticipated xenotransplant clinical trials.
Collapse
Affiliation(s)
| | - Andy Tully
- Program in Cardiac Xenotransplantation, Department of Surgery, University of Maryland, Baltimore, MD, USA
| | - Javier Galindo
- Program in Cardiac Xenotransplantation, Department of Surgery, University of Maryland, Baltimore, MD, USA
| | | | - Avneesh K Singh
- Program in Cardiac Xenotransplantation, Department of Surgery, University of Maryland, Baltimore, MD, USA
| | - Muhammad M Mohiuddin
- Program in Cardiac Xenotransplantation, Department of Surgery, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
24
|
Anand RP, Layer JV, Heja D, Hirose T, Lassiter G, Firl DJ, Paragas VB, Akkad A, Chhangawala S, Colvin RB, Ernst RJ, Esch N, Getchell K, Griffin AK, Guo X, Hall KC, Hamilton P, Kalekar LA, Kan Y, Karadagi A, Li F, Low SC, Matheson R, Nehring C, Otsuka R, Pandelakis M, Policastro RA, Pols R, Queiroz L, Rosales IA, Serkin WT, Stiede K, Tomosugi T, Xue Y, Zentner GE, Angeles-Albores D, Chris Chao J, Crabtree JN, Harken S, Hinkle N, Lemos T, Li M, Pantano L, Stevens D, Subedar OD, Tan X, Yin S, Anwar IJ, Aufhauser D, Capuano S, Kaufman DB, Knechtle SJ, Kwun J, Shanmuganayagam D, Markmann JF, Church GM, Curtis M, Kawai T, Youd ME, Qin W. Design and testing of a humanized porcine donor for xenotransplantation. Nature 2023; 622:393-401. [PMID: 37821590 PMCID: PMC10567564 DOI: 10.1038/s41586-023-06594-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 08/31/2023] [Indexed: 10/13/2023]
Abstract
Recent human decedent model studies1,2 and compassionate xenograft use3 have explored the promise of porcine organs for human transplantation. To proceed to human studies, a clinically ready porcine donor must be engineered and its xenograft successfully tested in nonhuman primates. Here we describe the design, creation and long-term life-supporting function of kidney grafts from a genetically engineered porcine donor transplanted into a cynomolgus monkey model. The porcine donor was engineered to carry 69 genomic edits, eliminating glycan antigens, overexpressing human transgenes and inactivating porcine endogenous retroviruses. In vitro functional analyses showed that the edited kidney endothelial cells modulated inflammation to an extent that was indistinguishable from that of human endothelial cells, suggesting that these edited cells acquired a high level of human immune compatibility. When transplanted into cynomolgus monkeys, the kidneys with three glycan antigen knockouts alone experienced poor graft survival, whereas those with glycan antigen knockouts and human transgene expression demonstrated significantly longer survival time, suggesting the benefit of human transgene expression in vivo. These results show that preclinical studies of renal xenotransplantation could be successfully conducted in nonhuman primates and bring us closer to clinical trials of genetically engineered porcine renal grafts.
Collapse
Affiliation(s)
| | | | | | - Takayuki Hirose
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Grace Lassiter
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel J Firl
- eGenesis, Cambridge, MA, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | - Ahmad Karadagi
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Feng Li
- eGenesis, Cambridge, MA, USA
| | | | - Rudy Matheson
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ryo Otsuka
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Toshihide Tomosugi
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Imran J Anwar
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - David Aufhauser
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Saverio Capuano
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Dixon B Kaufman
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Stuart J Knechtle
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - James F Markmann
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | | | - Tatsuo Kawai
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
25
|
Loupy A, Goutaudier V, Giarraputo A, Mezine F, Morgand E, Robin B, Khalil K, Mehta S, Keating B, Dandro A, Certain A, Tharaux PL, Narula N, Tissier R, Giraud S, Hauet T, Pass HI, Sannier A, Wu M, Griesemer A, Ayares D, Tatapudi V, Stern J, Lefaucheur C, Bruneval P, Mangiola M, Montgomery RA. Immune response after pig-to-human kidney xenotransplantation: a multimodal phenotyping study. Lancet 2023; 402:1158-1169. [PMID: 37598688 DOI: 10.1016/s0140-6736(23)01349-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Cross-species immunological incompatibilities have hampered pig-to-human xenotransplantation, but porcine genome engineering recently enabled the first successful experiments. However, little is known about the immune response after the transplantation of pig kidneys to human recipients. We aimed to precisely characterise the early immune responses to the xenotransplantation using a multimodal deep phenotyping approach. METHODS We did a complete phenotyping of two pig kidney xenografts transplanted to decedent humans. We used a multimodal strategy combining morphological evaluation, immunophenotyping (IgM, IgG, C4d, CD68, CD15, NKp46, CD3, CD20, and von Willebrand factor), gene expression profiling, and whole-transcriptome digital spatial profiling and cell deconvolution. Xenografts before implantation, wild-type pig kidney autografts, as well as wild-type, non-transplanted pig kidneys with and without ischaemia-reperfusion were used as controls. FINDINGS The data collected from xenografts suggested early signs of antibody-mediated rejection, characterised by microvascular inflammation with immune deposits, endothelial cell activation, and positive xenoreactive crossmatches. Capillary inflammation was mainly composed of intravascular CD68+ and CD15+ innate immune cells, as well as NKp46+ cells. Both xenografts showed increased expression of genes biologically related to a humoral response, including monocyte and macrophage activation, natural killer cell burden, endothelial activation, complement activation, and T-cell development. Whole-transcriptome digital spatial profiling showed that antibody-mediated injury was mainly located in the glomeruli of the xenografts, with significant enrichment of transcripts associated with monocytes, macrophages, neutrophils, and natural killer cells. This phenotype was not observed in control pig kidney autografts or in ischaemia-reperfusion models. INTERPRETATION Despite favourable short-term outcomes and absence of hyperacute injuries, our findings suggest that antibody-mediated rejection in pig-to-human kidney xenografts might be occurring. Our results suggest specific therapeutic targets towards the humoral arm of rejection to improve xenotransplantation results. FUNDING OrganX and MSD Avenir.
Collapse
Affiliation(s)
- Alexandre Loupy
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.
| | - Valentin Goutaudier
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alessia Giarraputo
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Cardiovascular Pathology and Pathological Anatomy, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Fariza Mezine
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Erwan Morgand
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Blaise Robin
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Pharmacy, NYU Langone Health, New York, NY, USA
| | - Sapna Mehta
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Brendan Keating
- Division of Transplantation, Department of Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | | | - Anaïs Certain
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center, PARCC, INSERM U970, Université Paris Cité, Paris, France
| | - Navneet Narula
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Renaud Tissier
- Ecole Nationale Vétérinaire d'Alfort, IMRB, After ROSC Network, Maisons-Alfort, France
| | - Sébastien Giraud
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, Poitiers, France
| | - Thierry Hauet
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, Poitiers, France
| | - Harvey I Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, NY, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Aurélie Sannier
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Department of Pathology, Bichat Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Ming Wu
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Carmen Lefaucheur
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Patrick Bruneval
- Université Paris Cité, INSERM U970 PARCC, Paris Institute for Transplantation and Organ Regeneration, Paris, France; Department of Pathology, Georges Pompidou European Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
26
|
Chornenkyy Y, Yamamoto T, Hara H, Stowell SR, Ghiran I, Robson SC, Cooper DKC. Future prospects for the clinical transfusion of pig red blood cells. Blood Rev 2023; 61:101113. [PMID: 37474379 PMCID: PMC10968389 DOI: 10.1016/j.blre.2023.101113] [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: 04/24/2023] [Revised: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Transfusion of allogeneic human red blood cell (hRBCs) is limited by supply and compatibility between individual donors and recipients. In situations where the blood supply is constrained or when no compatible RBCs are available, patients suffer. As a result, alternatives to hRBCs that complement existing RBC transfusion strategies are needed. Pig RBCs (pRBCs) could provide an alternative because of their abundant supply, and functional similarities to hRBCs. The ability to genetically modify pigs to limit pRBC immunogenicity and augment expression of human 'protective' proteins has provided major boosts to this research and opens up new therapeutic avenues. Although deletion of expression of xenoantigens has been achieved in genetically-engineered pigs, novel genetic methods are needed to introduce human 'protective' transgenes into pRBCs at the high levels required to prevent hemolysis and extend RBC survival in vivo. This review addresses recent progress and examines future prospects for clinical xenogeneic pRBC transfusion.
Collapse
Affiliation(s)
- Yevgen Chornenkyy
- Department of Pathology, McGaw Medical Center of Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Takayuki Yamamoto
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA; Division of Transplantation, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - David K C Cooper
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Swatek AM, Parekh KR. Lung Xenotransplantation. Thorac Surg Clin 2023; 33:291-297. [PMID: 37414485 DOI: 10.1016/j.thorsurg.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Although efforts have been made to expand the pool of donor lung allografts for human lung transplantation, a shortage remains. Lung xenotransplantation has been proposed as an alternative approach, but lung xenotransplantation in humans has not yet been reported. In addition, significant biological and ethical barriers will have to be addressed before clinical trials can be undertaken. However, significant progress has been made toward addressing biological incompatibilities that present a barrier, and recent advances in genetic engineering tools promise to accelerate further progress.
Collapse
Affiliation(s)
- Anthony M Swatek
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, SE500GH, Iowa City, IA 52242, USA
| | - Kalpaj R Parekh
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, SE500GH, Iowa City, IA 52242, USA.
| |
Collapse
|
28
|
Cross-Najafi AA, Farag K, Isidan A, Li W, Zhang W, Lin Z, Walsh JR, Lopez K, Park Y, Higgins NG, Cooper DK, Ekser B, Li P. Co-expression of HLA-E and HLA-G on genetically modified porcine endothelial cells attenuates human NK cell-mediated degranulation. Front Immunol 2023; 14:1217809. [PMID: 37529053 PMCID: PMC10387534 DOI: 10.3389/fimmu.2023.1217809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Natural killer (NK) cells play an important role in immune rejection in solid organ transplantation. To mitigate human NK cell activation in xenotransplantation, introducing inhibitory ligands on xenografts via genetic engineering of pigs may protect the graft from human NK cell-mediated cytotoxicity and ultimately improve xenograft survival. In this study, non-classical HLA class I molecules HLA-E and HLA-G were introduced in an immortalized porcine liver endothelial cell line with disruption of five genes (GGTA1, CMAH, β4galNT2, SLA-I α chain, and β-2 microglobulin) encoding three major carbohydrate xenoantigens (αGal, Neu5Gc, and Sda) and swine leukocyte antigen class I (SLA-I) molecules. Expression of HLA-E and/or HLA-G on pig cells were confirmed by flow cytometry. Endogenous HLA-G molecules as well as exogenous HLA-G VL9 peptide could dramatically enhance HLA-E expression on transfected pig cells. We found that co-expression of HLA-E and HLA-G on porcine cells led to a significant reduction in human NK cell activation compared to the cells expressing HLA-E or HLA-G alone and the parental cell line. NK cell activation was assessed by analysis of CD107a expression in CD3-CD56+ population gated from human peripheral blood mononuclear cells. CD107a is a sensitive marker of NK cell activation and correlates with NK cell degranulation and cytotoxicity. HLA-E and/or HLA-G on pig cells did not show reactivity to human sera IgG and IgM antibodies. This in vitro study demonstrated that co-expression of HLA-E and HLA-G on genetically modified porcine endothelial cells provided a superior inhibition in human xenoreactive NK cells, which may guide further genetic engineering of pigs to prevent human NK cell mediated rejection.
Collapse
Affiliation(s)
- Arthur A. Cross-Najafi
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kristine Farag
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Abdulkadir Isidan
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Wenjun Zhang
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zhansong Lin
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
| | - Julia R. Walsh
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kevin Lopez
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yujin Park
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nancy G. Higgins
- Transplant Immunology, Indiana University Health, Indianapolis, IN, United States
| | - David K.C. Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Cambridge, MA, United States
| | - Burcin Ekser
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ping Li
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
29
|
Habibabady Z, McGrath G, Kinoshita K, Maenaka A, Ikechukwu I, Elias GF, Zaletel T, Rosales I, Hara H, Pierson RN, Cooper DKC. Antibody-mediated rejection in xenotransplantation: Can it be prevented or reversed? Xenotransplantation 2023; 30:e12816. [PMID: 37548030 PMCID: PMC11101061 DOI: 10.1111/xen.12816] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Antibody-mediated rejection (AMR) is the commonest cause of failure of a pig graft after transplantation into an immunosuppressed nonhuman primate (NHP). The incidence of AMR compared to acute cellular rejection is much higher in xenotransplantation (46% vs. 7%) than in allotransplantation (3% vs. 63%) in NHPs. Although AMR in an allograft can often be reversed, to our knowledge there is no report of its successful reversal in a pig xenograft. As there is less experience in preventing or reversing AMR in models of xenotransplantation, the results of studies in patients with allografts provide more information. These include (i) depletion or neutralization of serum anti-donor antibodies, (ii) inhibition of complement activation, (iii) therapies targeting B or plasma cells, and (iv) anti-inflammatory therapy. Depletion or neutralization of anti-pig antibody, for example, by plasmapheresis, is effective in depleting antibodies, but they recover within days. IgG-degrading enzymes do not deplete IgM. Despite the expression of human complement-regulatory proteins on the pig graft, inhibition of systemic complement activation may be necessary, particularly if AMR is to be reversed. Potential therapies include (i) inhibition of complement activation (e.g., by IVIg, C1 INH, or an anti-C5 antibody), but some complement inhibitors are not effective in NHPs, for example, eculizumab. Possible B cell-targeted therapies include (i) B cell depletion, (ii) plasma cell depletion, (iii) modulation of B cell activation, and (iv) enhancing the generation of regulatory B and/or T cells. Among anti-inflammatory agents, anti-IL6R mAb and TNF blockers are increasingly being tested in xenotransplantation models, but with no definitive evidence that they reverse AMR. Increasing attention should be directed toward testing combinations of the above therapies. We suggest that treatment with a systemic complement inhibitor is likely to be most effective, possibly combined with anti-inflammatory agents (if these are not already being administered). Ultimately, it may require further genetic engineering of the organ-source pig to resolve the problem entirely, for example, knockout or knockdown of SLA, and/or expression of PD-L1, HLA E, and/or HLA-G.
Collapse
Affiliation(s)
- Zahra Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ileka Ikechukwu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela F. Elias
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Tjasa Zaletel
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Hara
- Yunnan Xenotransplantation Engineering Research Center, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Xi J, Zheng W, Chen M, Zou Q, Tang C, Zhou X. Genetically engineered pigs for xenotransplantation: Hopes and challenges. Front Cell Dev Biol 2023; 10:1093534. [PMID: 36712969 PMCID: PMC9878146 DOI: 10.3389/fcell.2022.1093534] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/31/2022] [Indexed: 01/14/2023] Open
Abstract
The shortage of donor resources has greatly limited the application of clinical xenotransplantation. As such, genetically engineered pigs are expected to be an ideal organ source for xenotransplantation. Most current studies mainly focus on genetically modifying organs or tissues from donor pigs to reduce or prevent attack by the human immune system. Another potential organ source is interspecies chimeras. In this paper, we reviewed the progress of the genetically engineered pigs from the view of immunologic barriers and strategies, and discussed the possibility and challenges of the interspecies chimeras.
Collapse
|
31
|
Lei T, Chen L, Wang K, Du S, Gonelle-Gispert C, Wang Y, Buhler LH. Genetic engineering of pigs for xenotransplantation to overcome immune rejection and physiological incompatibilities: The first clinical steps. Front Immunol 2022; 13:1031185. [PMID: 36561750 PMCID: PMC9766364 DOI: 10.3389/fimmu.2022.1031185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Xenotransplantation has the potential to solve the shortfall of human organ donors. Genetically modified pigs have been considered as potential animal donors for human xenotransplantation and have been widely used in preclinical research. The genetic modifications aim to prevent the major species-specific barriers, which include humoral and cellular immune responses, and physiological incompatibilities such as complement and coagulation dysfunctions. Genetically modified pigs can be created by deleting several pig genes related to the synthesis of various pig specific antigens or by inserting human complement- and coagulation-regulatory transgenes. Finally, in order to reduce the risk of infection, genes related to porcine endogenous retroviruses can be knocked down. In this review, we focus on genetically modified pigs and comprehensively summarize the immunological mechanism of xenograft rejection and recent progress in preclinical and clinical studies. Overall, both genetically engineered pig-based xenografts and technological breakthroughs in the biomedical field provide a promising foundation for pig-to-human xenotransplantation in the future.
Collapse
Affiliation(s)
- Tiantian Lei
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Lin Chen
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Kejing Wang
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | | | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Leo H. Buhler
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
32
|
Lu TY, Xu XL, Du XG, Wei JH, Yu JN, Deng SL, Qin C. Advances in Innate Immunity to Overcome Immune Rejection during Xenotransplantation. Cells 2022; 11:cells11233865. [PMID: 36497122 PMCID: PMC9735653 DOI: 10.3390/cells11233865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Transplantation is an effective approach for treating end-stage organ failure. There has been a long-standing interest in xenotransplantation as a means of increasing the number of available organs. In the past decade, there has been tremendous progress in xenotransplantation accelerated by the development of rapid gene-editing tools and immunosuppressive therapy. Recently, the heart and kidney from pigs were transplanted into the recipients, which suggests that xenotransplantation has entered a new era. The genetic discrepancy and molecular incompatibility between pigs and primates results in barriers to xenotransplantation. An increasing body of evidence suggests that innate immune responses play an important role in all aspects of the xenogeneic rejection. Simultaneously, the role of important cellular components like macrophages, natural killer (NK) cells, and neutrophils, suggests that the innate immune response in the xenogeneic rejection should not be underestimated. Here, we summarize the current knowledge about the innate immune system in xenotransplantation and highlight the key issues for future investigations. A better understanding of the innate immune responses in xenotransplantation may help to control the xenograft rejection and design optimal combination therapies.
Collapse
Affiliation(s)
- Tian-Yu Lu
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of animal model, Beijing 100021, China
| | - Xue-Ling Xu
- National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xu-Guang Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jin-Hua Wei
- Cardiovascular Surgery Department, Center of Laboratory Medicine, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jia-Nan Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of animal model, Beijing 100021, China
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of animal model, Beijing 100021, China
- Correspondence: (S.-L.D.); (C.Q.)
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, National Human Diseases Animal Model Resource Center, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of animal model, Beijing 100021, China
- Changping National Laboratory (CPNL), Beijing 102206, China
- Correspondence: (S.-L.D.); (C.Q.)
| |
Collapse
|
33
|
Deng J, Yang L, Wang Z, Ouyang H, Yu H, Yuan H, Pang D. Advance of genetically modified pigs in xeno-transplantation. Front Cell Dev Biol 2022; 10:1033197. [PMID: 36299485 PMCID: PMC9590650 DOI: 10.3389/fcell.2022.1033197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
As the standard of living improves, chronic diseases and end-stage organ failure have been a regular occurrence in human beings. Organ transplantation has become one of the hopes in the fight against chronic diseases and end-stage organ failure. However, organs available for transplantation are far from sufficient to meet the demand, leading to a major organ shortage crisis. To solve this problem, researchers have turned to pigs as their target since pigs have many advantages as xenograft donors. Pigs are considered the ideal organ donor for human xenotransplantation, but direct transplantation of porcine organs to humans faces many obstacles, such as hyperacute rejection, acute humoral xenograft rejection, coagulation dysregulation, inflammatory response, coagulation dysregulation, and endogenous porcine retroviral infection. Many transgenic strategies have been developed to overcome these obstacles. This review provides an overview of current advances in genetically modified pigs for xenotransplantation. Future genetic engineering-based delivery of safe and effective organs and tissues for xenotransplantation remains our goal.
Collapse
Affiliation(s)
- Jiacheng Deng
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lin Yang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Ziru Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongming Yuan
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| |
Collapse
|
34
|
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: 65] [Impact Index Per Article: 21.7] [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
|
35
|
Stewart ZA. Xenotransplantation: The Contribution of CRISPR/Cas9 Gene Editing Technology. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00380-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
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: 25] [Impact Index Per Article: 8.3] [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
|
37
|
Kogata S, Lo PC, Maeda A, Okamatsu C, Sato K, Yamamoto R, Haneda T, Yoneyama T, Toyama C, Eguchi H, Masahata K, Kamiyama M, Okuyama H, Miyagawa S. Suppression of macrophage-mediated xenogeneic rejection by the ectopic expression of human CD177. Transpl Immunol 2022; 74:101663. [PMID: 35835297 DOI: 10.1016/j.trim.2022.101663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Cellular xenogeneic rejection by the innate immune system is a major immunological obstruction that needs to be overcome for the successful clinical use of xenografts. Our focus has been on macrophage-mediated xenogeneic rejection, since suppressing macrophage function has considerable potential for practical applications in the area of xenotransplantation. We report herein on an investigation of the suppressive effect of human CD177 (hCD177) against macrophage-mediated xenogeneic rejection. Wild type swine aortic endothelial cell (SEC) and an SEC transfectant with hCD177 (SEC/hCD177) were co-cultured with macrophages, and the degree of cytotoxicity was evaluated by WST-8 assays, and phagocytosis was examined using Calcein-AM labeling methods. The expression of anti/pro-inflammatory cytokines was evaluated by RT-qPCR and the phosphorylation of SHP-1 on macrophages in co-culture was evaluated by Western blotting. The result of cytotoxicity assays indicated that hCD177 suppressed M1 macrophage-mediated xenogeneic rejection (vs. SEC, p < 0.0001). Similarly, the result of phagocytosis assays indicated that hCD177 suppressed it (vs. SEC, p < 0.05). In addition, hCD177 significantly suppressed the expression of IL-1β, a pro-inflammatory cytokine, in M1 macrophages (vs. SEC, p < 0.01). Luciferase assays using THP1-Lucia NF-kB also showed a significant difference in NF-kB activation (vs. SEC, p < 0.001). In addition, hCD177 was found to induce the phosphorylation of SHP-1 in M1 macrophages (vs. SEC, p < 0.05). These findings indicate that hCD177 suppresses M1 macrophage-mediated xenogeneic rejection, at least in part via in the phosphorylation of SHP-1.
Collapse
Affiliation(s)
- Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Division of Pediatric Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Pei-Chi Lo
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuki Sato
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Haneda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohisa Yoneyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| |
Collapse
|
38
|
Hu M, Hawthorne WJ, Yi S, O’Connell PJ. Cellular Immune Responses in Islet Xenograft Rejection. Front Immunol 2022; 13:893985. [PMID: 35874735 PMCID: PMC9300897 DOI: 10.3389/fimmu.2022.893985] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Porcine islets surviving the acute injury caused by humoral rejection and IBMIR will be subjected to cellular xenograft rejection, which is predominately mediated by CD4+ T cells and is characterised by significant infiltration of macrophages, B cells and T cells (CD4+ and CD8+). Overall, the response is different compared to the alloimmune response and more difficult to suppress. Activation of CD4+ T cells is both by direct and indirect antigen presentation. After activation they recruit macrophages and direct B cell responses. Although they are less important than CD4+ T cells in islet xenograft rejection, macrophages are believed to be a major effector cell in this response. Rodent studies have shown that xenoantigen-primed and CD4+ T cell-activated macrophages were capable of recognition and rejection of pancreatic islet xenografts, and they destroyed a graft via the secretion of various proinflammatory mediators, including TNF-α, reactive oxygen and nitrogen species, and complement factors. B cells are an important mediator of islet xenograft rejection via xenoantigen presentation, priming effector T cells and producing xenospecific antibodies. Depletion and/or inhibition of B cells combined with suppressing T cells has been suggested as a promising strategy for induction of xeno-donor-specific T- and B-cell tolerance in islet xenotransplantation. Thus, strategies that expand the influence of regulatory T cells and inhibit and/or reduce macrophage and B cell responses are required for use in combination with clinical applicable immunosuppressive agents to achieve effective suppression of the T cell-initiated xenograft response.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Wayne J. Hawthorne
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Shounan Yi
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Philip J. O’Connell
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Philip J. O’Connell,
| |
Collapse
|
39
|
Zhang J, Xu Y, Zhang Y, Bossila EA, Shi M, Zhao Y. Bioinformatic analysis as a first step to predict the compatibility of hematopoiesis and immune system genes between humans and pigs. Xenotransplantation 2022; 29:e12764. [PMID: 35695327 DOI: 10.1111/xen.12764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022]
Abstract
The shortage of allogeneic donor organs leaves its supply far short of clinical need. There are great expectations on xenotransplantation, especially with pigs' organs. With the genetic modification of donor pigs, the rejection and cross-species transmission issues have now been widely addressed. However, research on the compatibility of genes between humans and pigs was limited. We performed a systematic screening analysis of predicted incompatible genes between humans and pigs, judged by low protein sequence similarities or different predicted protein domain compositions. By combining with gene set enrichment analysis, we screened out several key genes of hematopoiesis and the immune system with possible incompatibilities, which might be important for establishing chimera and xenotransplantation between humans and pigs. There were seven chemokine genes, including CCL1, CCL5, CCL24, CCL25, CCL28, CXCL12, and CXCL16, that exhibited limited similarity between humans and pigs (similarity < 0.8). Among hematopoiesis process-related genes, 15 genes of adhesion molecules, Notch ligands, and cytokine receptors exhibited differences between humans and pigs. In complement and coagulation cascades, 19 genes showed low similarity and 77 genes had different domain compositions between humans and pigs. Our study provides a good reference for further genetic modification of pigs, which might be beneficial for xenotransplantation.
Collapse
Affiliation(s)
- Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yingzi Zhang
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Elhusseny A Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo, Egypt
| | - Mingpu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Eisenson DL, Hisadome Y, Yamada K. Progress in Xenotransplantation: Immunologic Barriers, Advances in Gene Editing, and Successful Tolerance Induction Strategies in Pig-To-Primate Transplantation. Front Immunol 2022; 13:899657. [PMID: 35663933 PMCID: PMC9157571 DOI: 10.3389/fimmu.2022.899657] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Organ transplantation is the most effective treatment for end stage organ failure, but there are not enough organs to meet burgeoning demand. One potential solution to this organ shortage is xenotransplantation using pig tissues. Decades of progress in xenotransplantation, accelerated by the development of rapid genome editing tools, particularly the advent of CRISPR-Cas9 gene editing technologies, have enabled remarkable advances in kidney and heart xenotransplantation in pig-to-nonhuman primates. These breakthroughs in large animal preclinical models laid the foundation for three recent pig-to-human transplants by three different groups: two kidney xenografts in brain dead recipients deemed ineligible for transplant, and one heart xenograft in the first clinical grade study of pig-to-human transplantation. However, despite tremendous progress, recent data including the first clinical case suggest that gene-modification alone will not overcome all xenogeneic immunologic barriers, and thus an active and innovative immunologic strategy is required for successful xenotransplantation. This review highlights xenogeneic immunologic barriers, advances in gene editing, and tolerance-inducing strategies in pig-to-human xenotransplantation.
Collapse
Affiliation(s)
- Daniel L Eisenson
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States.,Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Yu Hisadome
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Kazuhiko Yamada
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States.,Department of Surgery, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
41
|
Jiang Z, Fu M, Zhu D, Wang X, Li N, Ren L, He J, Yang G. Genetically modified immunomodulatory cell-based biomaterials in tissue regeneration and engineering. Cytokine Growth Factor Rev 2022; 66:53-73. [PMID: 35690567 DOI: 10.1016/j.cytogfr.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
To date, the wide application of cell-based biomaterials in tissue engineering and regeneration is remarkably hampered by immune rejection. Reducing the immunogenicity of cell-based biomaterials has become the latest direction in biomaterial research. Recently, genetically modified cell-based biomaterials with immunomodulatory genes have become a feasible solution to the immunogenicity problem. In this review, recent advances and future challenges of genetically modified immunomodulatory cell-based biomaterials are elaborated, including fabrication approaches, mechanisms of common immunomodulatory genes, application and, more importantly, current preclinical and clinical advances. The fabrication approaches can be categorized into commonly used (e.g., virus transfection) and newly developed approaches. The immunomodulatory mechanisms of representative genes involve complicated cell signaling pathways and metabolic activities. Wide application in curing multiple end-term diseases and replacing lifelong immunosuppressive therapy in multiple cell and organ transplantation models is demonstrated. Most significantly, practices of genetically modified organ transplantation have been conducted on brain-dead human decedent and even on living patients after a series of experiments on nonhuman primates. Nevertheless, uncertain biosecurity, nonspecific effects and overlooked personalization of current genetically modified immunomodulatory cell-based biomaterials are shortcomings that remain to be overcome.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Xueting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Lingfei Ren
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
42
|
Abstract
To bridge the gap between organ demand and supply, xenotransplantation has long been considered as a realistic option for end-stage organ failure. Early this year this promise became reality for David Bennett Sr., the first patient whose own failing heart was replaced with a xeno-pig heart. To get here has been a rollercoaster ride of physiological hurdles seemingly impossible to overcome, technological breakthroughs and ethical and safety concerns. It started in 1984, with Stephanie Fae Beauclair, also known as baby Fae, receiving a baboon heart, which allowed her to survive for another 30 days. For ethical reasons primate work was soon abandoned in favour of the pig. But increased phylogenetic distance also brought with it an increased immunological incompatibility. It has been the development of ever more sophisticated genetic engineering tools, which brought down the physiological barriers, enabled humanisation of porcine organs and helped addressing safety concerns. This renewed the confidence in xenotransplantation, brought new funding opportunities and resulted finally in the first in human trial.
Collapse
Affiliation(s)
- Konrad Fischer
- Chair of Livestock Biotechnology, School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technical University of Munich, Munich, Germany.
| |
Collapse
|
43
|
Brom VC, Burger C, Wirtz DC, Schildberg FA. The Role of Immune Checkpoint Molecules on Macrophages in Cancer, Infection, and Autoimmune Pathologies. Front Immunol 2022; 13:837645. [PMID: 35418973 PMCID: PMC8995707 DOI: 10.3389/fimmu.2022.837645] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized immunotherapy against various cancers over the last decade. The use of checkpoint inhibitors results in remarkable re-activation of patients’ immune system, but is also associated with significant adverse events. In this review, we emphasize the importance of cell-type specificity in the context of immune checkpoint-based interventions and particularly focus on the relevance of macrophages. Immune checkpoint blockade alters the dynamic macrophage phenotypes and thereby substantially manipulates therapeutical outcome. Considering the macrophage-specific immune checkpoint biology, it seems feasible to ameliorate the situation of patients with severe side effects and even increase the probability of survival for non-responders to checkpoint inhibition. Apart from malignancies, investigating immune checkpoint molecules on macrophages has stimulated their fundamental characterization and use in other diseases as well, such as acute and chronic infections and autoimmune pathologies. Although the macrophage-specific effect of checkpoint molecules has been less studied so far, the current literature shows that a macrophage-centered blockade of immune checkpoints as well as a stimulation of their expression represents promising therapeutic avenues. Ultimately, the therapeutic potential of a macrophage-focused checkpoint therapy might be maximized by diagnostically assessing individual checkpoint expression levels on macrophages, thereby personalizing an effective treatment approach for each patient having cancer, infection, or autoimmune diseases.
Collapse
Affiliation(s)
- Victoria C Brom
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
44
|
Maeda A, Kogata S, Toyama C, Lo PC, Okamatsu C, Yamamoto R, Masahata K, Kamiyama M, Eguchi H, Watanabe M, Nagashima H, Okuyama H, Miyagawa S. The Innate Cellular Immune Response in Xenotransplantation. Front Immunol 2022; 13:858604. [PMID: 35418992 PMCID: PMC8995651 DOI: 10.3389/fimmu.2022.858604] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/23/2022] [Indexed: 01/02/2023] Open
Abstract
Xenotransplantation is very attractive strategy for addressing the shortage of donors. While hyper acute rejection (HAR) caused by natural antibodies and complement has been well defined, this is not the case for innate cellular xenogeneic rejection. An increasing body of evidence suggests that innate cellular immune responses contribute to xenogeneic rejection. Various molecular incompatibilities between receptors and their ligands across different species typically have an impact on graft outcome. NK cells are activated by direct interaction as well as by antigen dependent cellular cytotoxicity (ADCC) mechanisms. Macrophages are activated through various mechanisms in xenogeneic conditions. Macrophages recognize CD47 as a "marker of self" through binding to SIRPα. A number of studies have shown that incompatibility of porcine CD47 against human SIRPα contributes to the rejection of xenogeneic target cells by macrophages. Neutrophils are an early responder cell that infiltrates xenogeneic grafts. It has also been reported that neutrophil extracellular traps (NETs) activate macrophages as damage-associated pattern molecules (DAMPs). In this review, we summarize recent insights into innate cellular xenogeneic rejection.
Collapse
Affiliation(s)
- Akira Maeda
- Department of Promotion for Blood and Marrow Transplantation, Aichi Medical University School of Medicine, Nagakute, Japan.,Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Pei-Chi Lo
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masahito Watanabe
- International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroshi Nagashima
- International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan.,International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| |
Collapse
|
45
|
Miyagawa S, Maeda A, Toyama C, Kogata S, Okamatsu C, Yamamoto R, Masahata K, Kamiyama M, Eguchi H, Watanabe M, Nagashima H, Ikawa M, Matsunami K, Okuyama H. Aspects of the Complement System in New Era of Xenotransplantation. Front Immunol 2022; 13:860165. [PMID: 35493484 PMCID: PMC9046582 DOI: 10.3389/fimmu.2022.860165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/07/2022] [Indexed: 01/16/2023] Open
Abstract
After producing triple (Gal, H-D and Sda)-KO pigs, hyperacute rejection appeared to no longer be a problem. However, the origin of xeno-rejection continues to be a controversial topic, including small amounts of antibodies and subsequent activation of the graft endothelium, the complement recognition system and the coagulation systems. The complement is activated via the classical pathway by non-Gal/H-D/Sda antigens and by ischemia-reperfusion injury (IRI), via the alternative pathway, especially on islets, and via the lectin pathway. The complement system therefore is still an important recognition and effector mechanism in xeno-rejection. All complement regulatory proteins (CRPs) regulate complement activation in different manners. Therefore, to effectively protect xenografts against xeno-rejection, it would appear reasonable to employ not only one but several CRPs including anti-complement drugs. The further assessment of antigens continues to be an important issue in the area of clinical xenotransplantation. The above conclusions suggest that the expression of sufficient levels of human CRPs on Triple-KO grafts is necessary. Moreover, multilateral inhibition on local complement activation in the graft, together with the control of signals between macrophages and lymphocytes is required.
Collapse
Affiliation(s)
- Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- International Institute for Bio-Resource Research, Meiji University, Kanagawa, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- *Correspondence: Shuji Miyagawa,
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahito Watanabe
- International Institute for Bio-Resource Research, Meiji University, Kanagawa, Japan
| | - Hiroshi Nagashima
- International Institute for Bio-Resource Research, Meiji University, Kanagawa, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Katsuyoshi Matsunami
- Department of Pharmacognosy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
46
|
Li T, Song R, Sun F, Saeed M, Guo X, Ye J, Chen F, Hou B, Zhu Q, Wang Y, Xie C, Tang L, Xu Z, Xu H, Yu H. Bioinspired magnetic nanocomplexes amplifying STING activation of tumor-associated macrophages to potentiate cancer immunotherapy. NANO TODAY 2022; 43:101400. [DOI: 10.1016/j.nantod.2022.101400] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
47
|
Mohiuddin MM, Goerlich CE, Singh AK, Zhang T, Tatarov I, Lewis B, Sentz F, Hershfeld A, Braileanu G, Odonkor P, Strauss E, Williams B, Burke A, Hittman J, Bhutta A, Tabatabai A, Gupta A, Vaught T, Sorrells L, Kuravi K, Dandro A, Eyestone W, Kaczorowski DJ, Ayares D, Griffith BP. Progressive genetic modifications of porcine cardiac xenografts extend survival to 9 months. Xenotransplantation 2022; 29:e12744. [PMID: 35357044 DOI: 10.1111/xen.12744] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 01/04/2023]
Abstract
We report orthotopic (life-supporting) survival of genetically engineered porcine cardiac xenografts (with six gene modifications) for almost 9 months in baboon recipients. This work builds on our previously reported heterotopic cardiac xenograft (three gene modifications) survival up to 945 days with an anti-CD40 monoclonal antibody-based immunosuppression. In this current study, life-supporting xenografts containing multiple human complement regulatory, thromboregulatory, and anti-inflammatory proteins, in addition to growth hormone receptor knockout (KO) and carbohydrate antigen KOs, were transplanted in the baboons. Selective "multi-gene" xenografts demonstrate survival greater than 8 months without the requirement of adjunctive medications and without evidence of abnormal xenograft thickness or rejection. These data demonstrate that selective "multi-gene" modifications improve cardiac xenograft survival significantly and may be foundational for paving the way to bridge transplantation in humans.
Collapse
Affiliation(s)
- Muhammad M Mohiuddin
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Corbin E Goerlich
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Avneesh K Singh
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tianshu Zhang
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ivan Tatarov
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Billeta Lewis
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Faith Sentz
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alena Hershfeld
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gheorghe Braileanu
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Patrick Odonkor
- Department of Anesthesiology, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Erik Strauss
- Department of Anesthesiology, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brittney Williams
- Department of Anesthesiology, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Allen Burke
- Department of Pathology, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jamie Hittman
- Department of Pathology, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adnan Bhutta
- Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ali Tabatabai
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anuj Gupta
- Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Amy Dandro
- Revivicor, Inc., Blacksburg, Virginia, USA
| | | | - David J Kaczorowski
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Bartley P Griffith
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Miura S, Habibabady ZA, Pollok F, Connolly M, Pratts S, Dandro A, Sorrells L, Karavi K, Phelps C, Eyestone W, Ayares D, Burdorf L, Azimzadeh A, Pierson RN. Effects of human TFPI and CD47 expression and selectin and integrin inhibition during GalTKO.hCD46 pig lung perfusion with human blood. Xenotransplantation 2022; 29:e12725. [PMID: 35234315 PMCID: PMC10207735 DOI: 10.1111/xen.12725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Loss of barrier function when GalTKO.hCD46 porcine lungs are perfused with human blood is associated with coagulation pathway dysregulation, innate immune system activation, and rapid sequestration of human formed blood elements. Here, we evaluate whether genetic expression of human tissue factor pathway inhibitor (hTFPI) and human CD47 (hCD47), alone or with combined selectin and integrin adhesion pathway inhibitors, delays GalTKO.hCD46 porcine lung injury or modulates neutrophil and platelet sequestration. METHODS In a well-established paired ex vivo lung perfusion model, GalTKO.hCD46.hTFPI.hCD47 transgenic porcine lungs (hTFPI.hCD47, n = 7) were compared to GalTKO.hCD46 lungs (reference, n = 5). All lung donor pigs were treated with a thromboxane synthase inhibitor, anti-histamine, and anti-GPIb integrin-blocking Fab, and were pre-treated with Desmopressin. In both genotypes, one lung of each pair was additionally treated with PSGL-1 and GMI-1271 (P- and E-selectin) and IB4 (CD11b/18 integrin) adhesion inhibitors (n = 6 hTFPI.hCD47, n = 3 reference). RESULTS All except for two reference lungs did not fail within 480 min when experiments were electively terminated. Selectin and integrin adhesion inhibitors moderately attenuated initial pulmonary vascular resistance (PVR) elevation in hTFPI.hCD47 lungs. Neutrophil sequestration was significantly delayed during the early time points following reperfusion and terminal platelet activation was attenuated in association with lungs expressing hTFPI.hCD47, but additional adhesion pathway inhibitors did not show further effects with either lung genotype. CONCLUSION Expression of hTFPI.hCD47 on porcine lung may be useful as part of an integrated strategy to prevent neutrophil adhesion and platelet activation that are associated with xenograft injury. Additionally, targeting canonical selectin and integrin adhesion pathways reduced PVR elevation associated with hTFPI.hCD47 expression, but did not significantly attenuate neutrophil or platelet sequestration. We conclude that other adhesive mechanisms mediate the residual sequestration of human formed blood elements to pig endothelium that occurs even in the context of the multiple genetic modifications and drug treatments tested here.
Collapse
Affiliation(s)
- Shuhei Miura
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Cardiovascular Surgery, Teine Keijinkai Hospital, Sapporo, Japan
| | - Zahra A. Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Franziska Pollok
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Margaret Connolly
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon Pratts
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | - Lars Burdorf
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Agnes Azimzadeh
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Podestà MA, Sykes M. Chimerism-Based Tolerance to Kidney Allografts in Humans: Novel Insights and Future Perspectives. Front Immunol 2022; 12:791725. [PMID: 35069574 PMCID: PMC8767096 DOI: 10.3389/fimmu.2021.791725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic rejection and immunosuppression-related toxicity severely affect long-term outcomes of kidney transplantation. The induction of transplantation tolerance – the lack of destructive immune responses to a transplanted organ in the absence of immunosuppression – could potentially overcome these limitations. Immune tolerance to kidney allografts from living donors has been successfully achieved in humans through clinical protocols based on chimerism induction with hematopoietic cell transplantation after non-myeloablative conditioning. Notably, two of these protocols have led to immune tolerance in a significant fraction of HLA-mismatched donor-recipient combinations, which represent the large majority of cases in clinical practice. Studies in mice and large animals have been critical in dissecting tolerance mechanisms and in selecting the most promising approaches for human translation. However, there are several key differences in tolerance induction between these models and humans, including the rate of success and stability of donor chimerism, as well as the relative contribution of different mechanisms in inducing donor-specific unresponsiveness. Kidney allograft tolerance achieved through durable full-donor chimerism may be due to central deletion of graft-reactive donor T cells, even though mechanistic data from patient series are lacking. On the other hand, immune tolerance attained with transient mixed chimerism-based protocols initially relies on Treg-mediated suppression, followed by peripheral deletion of donor-reactive recipient T-cell clones under antigenic pressure from the graft. These conclusions were supported by data deriving from novel high-throughput T-cell receptor sequencing approaches that allowed tracking of alloreactive repertoires over time. In this review, we summarize the most important mechanistic studies on tolerance induction with combined kidney-bone marrow transplantation in humans, discussing open issues that still need to be addressed and focusing on techniques developed in recent years to efficiently monitor the alloresponse in tolerance trials. These cutting-edge methods will be instrumental for the development of immune tolerance protocols with improved efficacy and to identify patients amenable to safe immunosuppression withdrawal.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milano, Italy
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Department of Surgery, Department of Microbiology and Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
50
|
Liang RY, Zhang KL, Chuang MH, Lin FH, Chen TC, Lin JN, Liang YJ, Li YA, Chen CH, Wong PLJ, Lin SZ, Lin PC. A One-Step, Monolayer Culture and Chemical-Based Approach to Generate Insulin-Producing Cells From Human Adipose-Derived Stem Cells to Mitigate Hyperglycemia in STZ-Induced Diabetic Rats. Cell Transplant 2022; 31:9636897221106995. [PMID: 36002988 PMCID: PMC9421045 DOI: 10.1177/09636897221106995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The global population of individuals afflicted with diabetes mellitus has been increasing year by year, and this disease poses a serious threat to human health as well as the economies worldwide. Pancreatic or islet transplantations provide one of the most effective and long-term therapies available to treat diabetes, but the scarcity and quality of pancreatic islets limit their use in treatments. Here, we report the development of a one-step, monolayer culture, and chemical-based protocol that efficiently mediates the differentiation of human adipose-derived stem cells (hADSCs) into insulin-producing cells (IPCs). Our data indicate that hADSCs in monolayer culture that are allowed to differentiate into IPCs are superior to those in suspension cultures with respect to insulin secretion capacity (213-fold increase), cell viability (93.5 ± 3.27% vs. 41.67 ± 13.17%), and response to glucose stimulation. Moreover, the expression of genes associated with pancreatic lineage specification, such as PDX1, ISL1, and INS (encoding insulin), were expressed at significantly higher levels during our differentiation protocol (6-fold for PDX1 and ISL1, 11.5-fold for INS). Importantly, in vivo studies demonstrated that transplantation with IPCs significantly mitigated hyperglycemia in streptozotocin-induced diabetic rats. Our results indicate that this one-step, rapid protocol increases the efficiency of IPC generation and that the chemical-based approach for IPC induction may reduce safety concerns associated with the use of IPCs for clinical applications, thereby providing a safe and effective cell-based treatment for diabetes.
Collapse
Affiliation(s)
- Ruei-Yue Liang
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
- Ruei-Yue Liang, Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan.
| | - Kai-Ling Zhang
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
| | - Ming-Hsi Chuang
- Department of Technology Management, Chung Hua University, Hsinchu, Taiwan
| | - Feng-Huei Lin
- Department of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Tzu-Chien Chen
- Department of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jhih-Ni Lin
- Department of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Jyun Liang
- Department of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-An Li
- Department of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Hung Chen
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
| | - Peggy Leh Jiunn Wong
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Tzu Chi Foundation, Hualien, Taiwan
- Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | - Po-Cheng Lin
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
| |
Collapse
|