1
|
Pan W, Zhang W, Zheng B, Camellato BR, Stern J, Lin Z, Khodadadi-Jamayran A, Kim J, Sommer P, Khalil K, Weldon E, Bai J, Zhu Y, Meyn P, Heguy A, Mangiola M, Griesemer A, Keating BJ, Montgomery RA, Xia B, Boeke JD. Cellular dynamics in pig-to-human kidney xenotransplantation. MED 2024; 5:1016-1029.e4. [PMID: 38776915 DOI: 10.1016/j.medj.2024.05.003] [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: 09/14/2023] [Revised: 01/30/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Xenotransplantation of genetically engineered porcine organs has the potential to address the challenge of organ donor shortage. Two cases of porcine-to-human kidney xenotransplantation were performed, yet the physiological effects on the xenografts and the recipients' immune responses remain largely uncharacterized. METHODS We performed single-cell RNA sequencing (scRNA-seq) and longitudinal RNA-seq analyses of the porcine kidneys to dissect xenotransplantation-associated cellular dynamics and xenograft-recipient interactions. We additionally performed longitudinal scRNA-seq of the peripheral blood mononuclear cells (PBMCs) to detect recipient immune responses across time. FINDINGS Although no hyperacute rejection signals were detected, scRNA-seq analyses of the xenografts found evidence of endothelial cell and immune response activation, indicating early signs of antibody-mediated rejection. Tracing the cells' species origin, we found human immune cell infiltration in both xenografts. Human transcripts in the longitudinal bulk RNA-seq revealed that human immune cell infiltration and the activation of interferon-gamma-induced chemokine expression occurred by 12 and 48 h post-xenotransplantation, respectively. Concordantly, longitudinal scRNA-seq of PBMCs also revealed two phases of the recipients' immune responses at 12 and 48-53 h. Lastly, we observed global expression signatures of xenotransplantation-associated kidney tissue damage in the xenografts. Surprisingly, we detected a rapid increase of proliferative cells in both xenografts, indicating the activation of the porcine tissue repair program. CONCLUSIONS Longitudinal and single-cell transcriptomic analyses of porcine kidneys and the recipient's PBMCs revealed time-resolved cellular dynamics of xenograft-recipient interactions during xenotransplantation. These cues can be leveraged for designing gene edits and immunosuppression regimens to optimize xenotransplantation outcomes. FUNDING This work was supported by NIH RM1HG009491 and DP5OD033430.
Collapse
Affiliation(s)
- Wanqing Pan
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Binghan Zheng
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brendan R Camellato
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ziyan Lin
- Applied Bioinformatics Laboratories (ABL), NYU Grossman School of Medicine, New York, NY 10016, USA
| | | | - Jacqueline Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Philip Sommer
- Department of Anesthesiology, Perioperative Care & Pain Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jiangshan Bai
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yinan Zhu
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Peter Meyn
- Genome Technology Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Heguy
- Genome Technology Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA; Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY 10016, USA; Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA.
| | - Bo Xia
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Society of Fellows, Harvard University, Cambridge, MA 02138, USA.
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
2
|
Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, Zhang W, Dowdell AK, Kim JI, Andrijevic D, Khalil K, Jaffe IS, Loza BL, Gragert L, Camellato BR, Oliveira MF, O'Brien DP, Chen HM, Weldon E, Gao H, Gandla D, Chang A, Bhatt R, Gao S, Lin X, Reddy KP, Kagermazova L, Habara AH, Widawsky S, Liang FX, Sall J, Loupy A, Heguy A, Taylor SEB, Zhu Y, Michael B, Jiang L, Jian R, Chong AS, Fairchild RL, Linna-Kuosmanen S, Kaikkonen MU, Tatapudi V, Lorber M, Ayares D, Mangiola M, Narula N, Moazami N, Pass H, Herati RS, Griesemer A, Kellis M, Snyder MP, Montgomery RA, Boeke JD, Keating BJ. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med 2024; 30:1448-1460. [PMID: 38760586 DOI: 10.1038/s41591-024-02972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Brian Piening
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Maedeh Mohebnasab
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University, Stanford, CA, 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
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Jacqueline I Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - David Andrijevic
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Ian S Jaffe
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Loren Gragert
- Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | - Han M Chen
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Chang
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Riyana Bhatt
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kriyana P Reddy
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alawi H Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sophie Widawsky
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Joseph Sall
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Alexandre Loupy
- Université Paris Cité, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Adriana Heguy
- Genome Technology Center, NYU Langone Health, New York, NY, USA
| | | | - Yinan Zhu
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Basil Michael
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - 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
| | - Nader Moazami
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Harvey Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Ramin S Herati
- Department of Medicine, 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
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 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
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA.
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA.
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Hansen-Estruch C, Cooper DK, Judd E. Physiological aspects of pig kidney xenotransplantation and implications for management following transplant. Xenotransplantation 2022; 29:e12743. [PMID: 35297098 PMCID: PMC9232961 DOI: 10.1111/xen.12743] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 02/19/2022] [Accepted: 03/03/2022] [Indexed: 12/16/2022]
Abstract
Successful organ transplantation between species is now possible, using genetic modifications. This article aims to provide a comprehensive overview of the differences and similarities in kidney function between humans, primates, and pigs, in preparation for pig-allograft to human xenotransplantation. The kidney, as the principal defender of body homeostasis, acts as a sensor, effector, and regulator of physiologic feedback systems. Considerations are made for anticipated effects on each system when a pig kidney is placed into a human recipient. Discussion topics include anatomy, global kidney function, sodium and water handling, kidney hormone production and response to circulating hormones, acid-base balance, and calcium and phosphorus handling. Based on available data, pig kidneys are anticipated to be compatible with human physiology, despite a few barriers.
Collapse
Affiliation(s)
- Christophe Hansen-Estruch
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K.C. Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric Judd
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
4
|
Yamamoto T, Hara H, Foote J, Wang L, Li Q, Klein EC, Schuurman HJ, Zhou H, Li J, Tector AJ, Zhang Z, Ezzelarab M, Lovingood R, Ayares D, Eckhoff DE, Cooper DKC, Iwase H. Life-supporting Kidney Xenotransplantation From Genetically Engineered Pigs in Baboons: A Comparison of Two Immunosuppressive Regimens. Transplantation 2019; 103:2090-2104. [PMID: 31283686 DOI: 10.1097/tp.0000000000002796] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The aims of this study were to evaluate the efficacy of US Food and Drug Administration-approved drugs in genetically engineered pig-to-baboon kidney xenotransplantation and compare the results with those using an anti-CD40 monoclonal antibody (mAb)-based regimen. METHODS Ten life-supporting kidney transplants were carried out in baboons using α1,3-galactosyltransferase gene-knockout/CD46 pigs with various other genetic manipulations aimed at controlling coagulation dysregulation. Eight transplants resulted in informative data. Immunosuppressive therapy consisted of induction with antithymocyte globulin and anti-CD20mAb, and maintenance based on either (1) CTLA4-Ig and/or tacrolimus (+rapamycin or mycophenolate mofetil) (GroupA [US Food and Drug Administration-approved regimens], n = 4) or (2) anti-CD40mAb + rapamycin (GroupB, n = 4). All baboons received corticosteroids, interleukin-6R blockade, and tumor necrosis factor-α blockade. Baboons were followed by clinical and laboratory monitoring of kidney function, coagulation, and immune parameters. At euthanasia, morphological and immunohistochemical studies were performed on the kidney grafts. RESULTS The median survival in GroupB was 186 days (range 90-260), which was significantly longer than in GroupA; median 14 days (range 12-32) (P < 0.01). Only GroupA baboons developed consumptive coagulopathy and the histopathological features of thrombotic microangiopathic glomerulopathy and interstitial arterial vasculitis. CONCLUSIONS Recognizing that the pig donors in each group differed in some genetic modifications, these data indicate that maintenance immunosuppression including anti-CD40mAb may be important to prevent pig kidney graft failure.
Collapse
Affiliation(s)
- Takayuki Yamamoto
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Jeremy Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL
| | - Liaoran Wang
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
- Second Affiliated Hospital, University of South China, Hengyang City, Hunan, China
| | - Qi Li
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
- Second Affiliated Hospital, University of South China, Hengyang City, Hunan, China
| | - Edwin C Klein
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA
| | | | - Hongmin Zhou
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
- Department of Cardiothoracic Surgery, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Li
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
- Second Affiliated Hospital, University of South China, Hengyang City, Hunan, China
| | - A Joseph Tector
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Zhongqiang Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
- Department of General Surgery and Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Ray Lovingood
- Kirklin Clinic Pharmacy, University of Alabama at Birmingham, Birmingham, AL
| | | | - Devin E Eckhoff
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
5
|
Kim SC, Mathews DV, Breeden CP, Higginbotham LB, Ladowski J, Martens G, Stephenson A, Farris AB, Strobert EA, Jenkins J, Walters EM, Larsen CP, Tector M, Tector AJ, Adams AB. Long-term survival of pig-to-rhesus macaque renal xenografts is dependent on CD4 T cell depletion. Am J Transplant 2019; 19:2174-2185. [PMID: 30821922 PMCID: PMC6658347 DOI: 10.1111/ajt.15329] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 01/22/2019] [Accepted: 02/04/2019] [Indexed: 01/25/2023]
Abstract
The shortage of available organs remains the greatest barrier to expanding access to transplant. Despite advances in genetic editing and immunosuppression, survival in experimental models of kidney xenotransplant has generally been limited to <100 days. We found that pretransplant selection of recipients with low titers of anti-pig antibodies significantly improved survival in a pig-to-rhesus macaque kidney transplant model (6 days vs median survival time 235 days). Immunosuppression included transient pan-T cell depletion and an anti-CD154-based maintenance regimen. Selective depletion of CD4+ T cells but not CD8+ T cells resulted in long-term survival (median survival time >400 days vs 6 days). These studies suggested that CD4+ T cells may have a more prominent role in xenograft rejection compared with CD8+ T cells. Although animals that received selective depletion of CD8+ T cells showed signs of early cellular rejection (marked CD4+ infiltrates), animals receiving selective CD4+ depletion exhibited normal biopsy results until late, when signs of chronic antibody rejection were present. In vitro study results suggested that rhesus CD4+ T cells required the presence of SLA class II to mount an effective proliferative response. The combination of low pretransplant anti-pig antibody and CD4 depletion resulted in consistent, long-term xenograft survival.
Collapse
Affiliation(s)
- SC Kim
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - DV Mathews
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - CP Breeden
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - LB Higginbotham
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - J Ladowski
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - G Martens
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - A Stephenson
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - AB Farris
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia
| | - EA Strobert
- Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - J Jenkins
- Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - EM Walters
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - CP Larsen
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia,Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| | - M Tector
- Comprehensive Transplant Institute, University of Alabama Birmingham School of Medicine, Birmingham, Alabama
| | - AJ Tector
- Comprehensive Transplant Institute, University of Alabama Birmingham School of Medicine, Birmingham, Alabama
| | - AB Adams
- Emory Transplant Center, Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia,Yerkes National Primate Research Center, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
6
|
In vivo regeneration of interspecies chimeric kidneys using a nephron progenitor cell replacement system. Sci Rep 2019; 9:6965. [PMID: 31061458 PMCID: PMC6502858 DOI: 10.1038/s41598-019-43482-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
Kidney regeneration is expected to be a new alternative treatment to the currently limited treatments for chronic kidney disease. By transplanting exogeneous nephron progenitor cells (NPCs) into the metanephric mesenchyme of a xenogeneic foetus, we aimed to regenerate neo-kidneys that originate from transplanted NPCs. Previously, we generated a transgenic mouse model enabling drug-induced ablation of NPCs (the Six2-iDTR mouse). We demonstrated that eliminating existing native host NPCs allowed their 100% replacement with donor mouse or rat NPCs, which could generate neo-nephrons on a culture dish. To apply this method to humans in the future, we examined the possibility of the in vivo regeneration of nephrons between different species via NPC replacement. We injected NPCs-containing rat renal progenitor cells and diphtheria toxin below the renal capsule of E13.5 metanephroi (MNs) of Six2-iDTR mice; the injected MNs were then transplanted into recipient rats treated with immunosuppressants. Consequently, we successfully regenerated rat/mouse chimeric kidneys in recipient rats receiving the optimal immunosuppressive therapy. We revealed a functional connection between the neo-glomeruli and host vessels and proper neo-glomeruli filtration. In conclusion, we successfully regenerated interspecies kidneys in vivo that acquired a vascular system. This novel strategy may represent an effective method for human kidney regeneration.
Collapse
|
7
|
Rivard CJ, Tanabe T, Lanaspa MA, Watanabe H, Nomura S, Andres-Hernando A, Garth K, Sekijima M, Ishimoto T, Ariyoshi Y, Garcia GE, Shah J, Lennan B, Tasaki M, Pomposelli T, Shimizu A, Sachs DH, Johnson RJ, Yamada K. Upregulation of CD80 on glomerular podocytes plays an important role in development of proteinuria following pig-to-baboon xeno-renal transplantation - an experimental study. Transpl Int 2018; 31:1164-1177. [PMID: 29722117 DOI: 10.1111/tri.13273] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/04/2018] [Accepted: 04/23/2018] [Indexed: 01/02/2023]
Abstract
We have previously reported that co-transplantation of the kidney with vascularized donor thymus from α-1,3-galactosyltransferase gene knockout pigs with an anti-CD154 with rituximab-based regimen led to improved xenograft survival in baboons with donor-specific unresponsiveness. However, nephrotic syndrome emerged as a complication in which the glomeruli showed mild mesangial expansion with similarities to minimal change disease (MCD) in humans. Since MCD is associated with CD80 expression in glomeruli and elevated urinary excretion, we evaluated a potential role for CD80 in xenograft nephropathy. Study 1 confirmed high urinary CD80 excretion in nephrotic animals with renal xenografts showing CD80 expression in glomeruli. In Study 2, baboons receiving xenografts received CTLA4-Ig once a week from the second postoperative week or no CTLA4-Ig. The non-CTLA4-Ig group developed severe proteinuria with modest mesangial expansion with high urinary excretion of CD80 and documented CD80 expression in glomerular podocytes. All of the recipients in non-CTLA4-Ig groups had to be euthanized before POD 60. In contrast, CTLA4-Ig group showed a marked reduction in proteinuria and survived significantly longer, up to 193 days. These results demonstrate that anti-CD80 targeted therapy represents a promising strategy for reduction of proteinuria following renal xeno-transplantation with improved survival.
Collapse
Affiliation(s)
- Christopher J Rivard
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Tatsu Tanabe
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Hironosuke Watanabe
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Shunichiro Nomura
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Krystle Garth
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Mitsuhiro Sekijima
- TBRC Laboratories, CTS, Massachusetts General Hospital, Charlestown, MA, USA
| | - Takuji Ishimoto
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Yuichi Ariyoshi
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Gabriela E Garcia
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Jigesh Shah
- TBRC Laboratories, CTS, Massachusetts General Hospital, Charlestown, MA, USA
| | - Boyd Lennan
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Masayuki Tasaki
- TBRC Laboratories, CTS, Massachusetts General Hospital, Charlestown, MA, USA
| | - Thomas Pomposelli
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Akira Shimizu
- TBRC Laboratories, CTS, Massachusetts General Hospital, Charlestown, MA, USA
| | - David H Sachs
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA.,TBRC Laboratories, CTS, Massachusetts General Hospital, Charlestown, MA, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Kazuhiko Yamada
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Remaining Physiological Barriers in Porcine Kidney Xenotransplantation: Potential Pathways behind Proteinuria as well as Factors Related to Growth Discrepancies following Pig-to-Kidney Xenotransplantation. J Immunol Res 2018; 2018:6413012. [PMID: 29687010 PMCID: PMC5857301 DOI: 10.1155/2018/6413012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/29/2017] [Accepted: 01/18/2018] [Indexed: 12/13/2022] Open
Abstract
Considerable shortages in the supply of available organs continue to plague the field of solid organ transplantation. Despite changes in allocation, as well as the utilization of extended criteria and living donors, the number of patients waiting for organs continues to grow at an alarming pace. Xenotransplantation, cross-species solid organ transplantation, offers one potential solution to this dilemma. Previous extensive research dedicated to this field has allowed for resolution of xenograft failure due to acute rejection, leaving new areas of unresolved challenges as barriers to success in large animal models. Specific to kidney xenotransplantation, recent data seems to indicate that graft compromise can occur due to discrepancies in growth between breeds of donors and significant proteinuria leading to nephrotic syndrome in the recipient. Given these potential limitations, herein, we review potential pathways behind proteinuria, as well as potential causative factors related to growth discrepancies. Control of both of these has the potential to allow xenotransplantation to become clinically applicable in an effort to resolve this organ shortage crisis.
Collapse
|
9
|
Yamada K, Shah JA, Tanabe T, Lanaspa MA, Johnson RJ. Xenotransplantation: Where Are We with Potential Kidney Recipients? Recent Progress and Potential Future Clinical Trials. CURRENT TRANSPLANTATION REPORTS 2017; 4:101-109. [PMID: 28989853 DOI: 10.1007/s40472-017-0149-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Inter-species transplantation, xenotransplantation, is becoming a realistic strategy to solve the organ shortage crisis. Here we focus on seminal publications that have driven research in xenotransplantation, as well as recently published literature and future endeavors. RECENT FINDINGS Advances in gene editing technology have allowed for the efficient production of multi-transgenic porcine donors leading improved xenograft survival in baboons, up to 2-years following heterotopic heart xenotransplantation and from weeks to several months following life-supporting kidney xenotransplanation. As technology evolves, additional challenges have arisen, including the development of proteinuria, early graft loss associated with porcine CMV, disparities in organ growth between donors and recipients as well as high-dose continuous immunosuppression requirements. To address these issues, our laboratory developed a tolerance-inducing protocol which has allowed for >6 months survival of a life-supporting kidney with further approaches currently underway to address the challenges mentioned above. SUMMARY Our recent findings, reviewed in this article, led us to develop methods to overcome obstacles, which, in conjunction with the work of others, are promising for future clinical applications of xenotransplantation.
Collapse
Affiliation(s)
- Kazuhiko Yamada
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Jigesh A Shah
- Transplantation Biology Research Laboratories, Massachusetts general Hospital, Harvard Medical School, Boston, MA
| | - Tatsu Tanabe
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora CO
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora CO
| |
Collapse
|
10
|
Results of life-supporting galactosyltransferase knockout kidneys in cynomolgus monkeys using two different sources of galactosyltransferase knockout Swine. Transplantation 2014; 98:419-26. [PMID: 25243512 DOI: 10.1097/tp.0000000000000314] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Various durations of survival have been observed in the xenotransplantation of life-supporting α-1,3-galactosyltransferase knockout (GalT-KO) porcine kidneys into nonhuman primates. Although others have demonstrated loss of GalT-KO-transplanted kidneys within 2 weeks, we have reported an average survival of 51 days with the cotransplantation of the kidney and vascularized thymus and an average of 29 days with the kidney alone. To determine the factors responsible for this difference in survival time, we performed xenogeneic kidney transplantations into cynomolgus monkeys with an anti-CD40L-based regimen using two different strains of GalT-KO swine, one derived from MGH miniature swine and the other obtained from Meji University. MATERIALS AND METHODS Eight cynomolgus moneys received GalT-KO kidneys. Three kidney grafts were from Massachusetts General Hospital (MGH)-Nippon Institute for Biological Science (NIBS) GalT-KO pigs and five GalT-KO grafts were from MEIJI GalT-KO swine. All cynomolgus recipients were treated identically. RESULTS Recipients of kidneys from the MGH GalT-KO kidneys swine, produced by nuclear transfer in Japan, survived an average of 28.7 days, whereas recipients of MEIJI GalT-KO kidneys swine survived an average of 9.2 days. Among the differences between these two groups, one potentially revealing disparity was that the MEIJI swine were positive for porcine cytomegalovirus, whereas the MGH-derived swine were negative. CONCLUSION This is the first study comparing renal xenotransplantation from two different sources of GalT-KO swine into nonhuman primates at a single center. The results demonstrate that porcine cytomegalovirus may be responsible for early loss of GalT-KO swine kidney xenografts.
Collapse
|
11
|
Cooper DKC, Satyananda V, Ekser B, van der Windt DJ, Hara H, Ezzelarab MB, Schuurman HJ. Progress in pig-to-non-human primate transplantation models (1998-2013): a comprehensive review of the literature. Xenotransplantation 2014; 21:397-419. [PMID: 25176336 DOI: 10.1111/xen.12127] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/03/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pig-to-non-human primate model is the standard choice for in vivo studies of organ and cell xenotransplantation. In 1998, Lambrigts and his colleagues surveyed the entire world literature and reported all experimental studies in this model. With the increasing number of genetically engineered pigs that have become available during the past few years, this model is being utilized ever more frequently. METHODS We have now reviewed the literature again and have compiled the data we have been able to find for the period January 1, 1998 to December 31, 2013, a period of 16 yr. RESULTS The data are presented for transplants of the heart (heterotopic and orthotopic), kidney, liver, lung, islets, neuronal cells, hepatocytes, corneas, artery patches, and skin. Heart, kidney, and, particularly, islet xenograft survival have increased significantly since 1998. DISCUSSION The reasons for this are briefly discussed. A comment on the limitations of the model has been made, particularly with regard to those that will affect progression of xenotransplantation toward the clinic.
Collapse
Affiliation(s)
- David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Schneider MKJ, Seebach JD. Xenotransplantation literature update, November-December 2013. Xenotransplantation 2014; 21:91-5. [PMID: 24444051 DOI: 10.1111/xen.12084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Mårten K J Schneider
- Laboratory of Vascular Immunology, Division of Internal Medicine, University Hospital Zurich, Switzerland
| | | |
Collapse
|