1
|
Krishnan A, Elde S, Ruaengsri C, Guenthart BA, Zhu Y, Fawad M, Lee A, Currie M, Ma MR, Hiesinger W, Shudo Y, MacArthur JW, Woo YJ. Survival, function, and immune profiling after beating heart transplantation. J Thorac Cardiovasc Surg 2025; 169:924-931. [PMID: 39111693 DOI: 10.1016/j.jtcvs.2024.07.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/15/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE Ex vivo normothermic perfusion of cardiac allografts has expanded the donor pool for heart transplant. Using a beating heart implantation method avoids the second cardioplegic arrest and subsequent ischemia-reperfusion injury typically associated with ex vivo heart perfusion. We sought to describe our institutional experience with beating heart transplantation. METHODS This was a single-institution retrospective study of adult patients who underwent heart transplantation using ex vivo heart perfusion (EVHP) and a beating heart implantation technique between October 2022 and March 2024. Primary outcomes of interest included survival, initiation of mechanical circulatory support, and rejection. A subanalysis of our institutional series of nonbeating deceased after circulatory death (DCD) heart transplantations was performed as well. RESULTS Twenty-four patients underwent isolated heart transplantation with the use of ex vivo heart perfusion and beating heart implantation between October 2022 and March 2024; 21 (87.5%) received hearts from DCD donors, and 3 (12.5%) received hearts from deceased after brain death (DBD) donors. The median duration of follow-up was 192 days (interquartile range [IQR], 124-253.5 days), and 23 out of 24 patients (95.8%) were alive at last follow-up. No patients required initiation of mechanical circulatory support. The majority of patients had pathologic grade 0 rejection at the time of biopsy (n = 16; 66.7%), and the median cell-free DNA percent was 0.04% (IQR, 0.04%-0.09%). The rate of mechanical circulatory support initiation in the 22-patient nonbeating DCD heart transplant cohort was significantly higher, at 36.4% (P < .005). CONCLUSIONS A beating heart implantation technique can be used for transplantation of DCD and DBD hearts on EVHP and is associated with excellent survival and low levels of rejection.
Collapse
Affiliation(s)
- Aravind Krishnan
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Chawannuch Ruaengsri
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Brandon A Guenthart
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Moeed Fawad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Anson Lee
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Maria Currie
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Michael R Ma
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - John Ward MacArthur
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif; Department of Bioengineering, Stanford University School of Engineering, Stanford, Calif.
| |
Collapse
|
2
|
Kanazawa R, Goto R, Harada T, Ota T, Kobayashi N, Shibuya K, Ganchiku Y, Watanabe M, Zaitsu M, Kawamura N, Shimamura T, Taketomi A. Early graft-infiltrating lymphocytes are not associated with graft rejection in a mouse model of skin transplantation. Scand J Immunol 2024; 100:e13397. [PMID: 39080853 DOI: 10.1111/sji.13397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 11/05/2024]
Abstract
Graft-infiltrating lymphocytes (GILs) play an important role in promoting rejection after organ transplantation. We recently reported that GILs that accumulated up to 3 days post-transplantation did not promote rejection, whereas GILs present 3-5 days post-transplantation promoted rejection in a mouse heart transplantation model. However, the immunological behaviour of GILs in murine skin transplantation remains unclear. GILs were isolated on days 3, 5 or 7 post-transplantation from C57BL/6 (B6) allogeneic skin grafts transplanted onto BALB/c mice. BALB/c Rag2-/- γc-/- mice (BRGs) underwent B6 skin graft transplantation 10 weeks after adoptive transfer of day 3, 5, or 7 GILs. BRGs reconstituted with day 5 or 7 GILs completely rejected B6 grafts. However, when B6 grafts harvested from recipient BALB/c mice on day 5 or 7 were re-transplanted into BRGs, half of the re-transplanted day 5 grafts established long-term survival, although all re-transplanted day 7 grafts were rejected. BRGs reconstituted with day 3 GILs did not reject B6 grafts. Consistently, re-transplantation using day 3 skin grafts resulted in no rejection. Administration of anti-CD25 antibodies did not prevent the phenomenon observed for the day 3 skin grafts. Furthermore, BRGs reconstituted with splenocytes from naïve BALB/c mice immediately rejected the naïve B6 skin grafts and the re-transplanted day 3 B6 grafts, suggesting that day 3 GILs were unable to induce allograft rejection during the rejection process. In conclusion, the immunological role of GILs depends on the time since transplantation. Day 3 GILs had neither protective nor alloreactive effects in the skin transplant model.
Collapse
Affiliation(s)
- Ryo Kanazawa
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Ryoichi Goto
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Takuya Harada
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Takuji Ota
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Nozomi Kobayashi
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Kazuaki Shibuya
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Yoshikazu Ganchiku
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Masaaki Watanabe
- Department of Transplant Surgery, Hokkaido University, Sapporo, Japan
| | - Masaaki Zaitsu
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
| | - Norio Kawamura
- Department of Transplant Surgery, Hokkaido University, Sapporo, Japan
| | - Tsuyoshi Shimamura
- Division of Organ Transplantation, Hokkaido University Hospital, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery 1, Hokkaido University, Sapporo, Japan
- Department of Transplant Surgery, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Lin S, Zeng H, Wang C, Chai Z, Zhang X, Yang B, Chi J, Zhang Y, Hu Z. Discovery of novel natural cardiomyocyte protectants from a toxigenic fungus Stachybotrys chartarum. Bioorg Chem 2024; 148:107461. [PMID: 38788363 DOI: 10.1016/j.bioorg.2024.107461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Stachybatranones A-F (1a/1b and 2-6) and three known analogues, namely methylatranones A and B (7 and 8) and atranone B (9), were isolated and identified from a toxigenic fungus Stachybotrys chartarum. Their structures and absolute configurations were elucidated via the extensive spectroscopic data, comparison of the experimental electronic circular dichroism (ECD) data, and single-crystal X-ray diffraction analyses. Structurally, compounds 2-6 belonged to a rare class of C-alkylated dolabellanes, featuring a unique five-membered hemiketal ring and a γ-butyrolactone moiety both fused to an 11-membered carbocyclic system, while compound 1 (1a/1b) represented the first example of a 5-11-6-fused atranone possessing a 2,3-butanediol moiety. The cardiomyocyte protective activity assay revealed that compounds 1-9 ameliorated cold ischemic injury at 24 h post cold ischemia (CI), with compounds 1 and 4 acting in a dose-dependent manner. Moreover, compound 1 prevented cold ischemia induced dephosphorylation of PI3K and AKT acting in a dose-dependent manner. In this study, a new class of natural products were found to protect cardiomyocytes against cold ischemic injury, providing a potential option for the development of novel cardioprotectants in heart transplant medicine.
Collapse
Affiliation(s)
- Shuang Lin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Hanxiao Zeng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Chenyang Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Zixue Chai
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xueke Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Beiye Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jiangyang Chi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, PR China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Zhengxi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
4
|
Franco-Acevedo A, Comes J, Mack JJ, Valenzuela NM. New insights into maladaptive vascular responses to donor specific HLA antibodies in organ transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1146040. [PMID: 38993843 PMCID: PMC11235244 DOI: 10.3389/frtra.2023.1146040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 07/13/2024]
Abstract
Transplant vasculopathy (TV) causes thickening of donor blood vessels in transplanted organs, and is a significant cause of graft loss and mortality in allograft recipients. It is known that patients with repeated acute rejection and/or donor specific antibodies are predisposed to TV. Nevertheless, the exact molecular mechanisms by which alloimmune injury culminates in this disease have not been fully delineated. As a result of this incomplete knowledge, there is currently a lack of effective therapies for this disease. The immediate intracellular signaling and the acute effects elicited by anti-donor HLA antibodies are well-described and continuing to be revealed in deeper detail. Further, advances in rejection diagnostics, including intragraft gene expression, provide clues to the inflammatory changes within allografts. However, mechanisms linking these events with long-term outcomes, particularly the maladaptive vascular remodeling seen in transplant vasculopathy, are still being delineated. New evidence demonstrates alterations in non-coding RNA profiles and the occurrence of endothelial to mesenchymal transition (EndMT) during acute antibody-mediated graft injury. EndMT is also readily apparent in numerous settings of non-transplant intimal hyperplasia, and lessons can be learned from advances in those fields. This review will provide an update on these recent developments and remaining questions in our understanding of HLA antibody-induced vascular damage, framed within a broader consideration of manifestations and implications across transplanted organ types.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Johanna Comes
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA, United States
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
5
|
Goto R, Fukasaku Y, Ganchiku Y, Kawamura N, Watanabe M, Ota T, Hatanaka KC, Suzuki T, Shimamura T, Taketomi A. Post-transplant donor-specific anti-HLA antibodies with a higher mean fluorescence intensity are associated with graft fibrosis in pediatric living donor liver transplantation. Front Pediatr 2023; 11:1172516. [PMID: 37181419 PMCID: PMC10168538 DOI: 10.3389/fped.2023.1172516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
The roles of post-transplant anti-HLA donor specific antibody (DSA) in pediatric liver transplantation (LT), including therapeutic strategies, remain controversial. This study aimed to identify the risks of post-transplant DSA for graft fibrosis progression in pediatric living donor LT (LDLT). We retrospectively evaluated 88 LDLT pediatric cases between December 1995 and November 2019. DSAs were assessed with single antigen bead test. Graft fibrosis was histopathologically scored with METAVIR and the centrilobular sinusoidal fibrosis system. Post-transplant DSAs were detected in 37 (52.9%) cases at 10.8 (1.3-26.9) years post-LDLT. The histopathological examination of 32 pediatric cases with post-transplant DSA revealed that 7 (21.9%) with a high DSA-MFI (≥9,378) showed graft fibrosis progression (≥F2). No graft fibrosis was observed in the subjects with a low DSA-MFI. The risk factors for developing graft fibrosis in pediatric cases with post-transplant DSA were an older graft age (>46.5 years old), lower platelet count (<10.7 × 104/ml) and higher Fib4 index (>0.7807, recipient age; >1.8952, donor age). Limited efficacy of additional immunosuppressants was observed in DSA positive pediatric cases. In conclusion, pediatric cases with a high DSA-MFI and risk factors should undergo a histological examination. The appropriate treatment for post-transplant DSA in pediatric LT needs to be determined.
Collapse
Affiliation(s)
- Ryoichi Goto
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasutomo Fukasaku
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshikazu Ganchiku
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Norio Kawamura
- Department of Transplant Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masaaki Watanabe
- Department of Transplant Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takuji Ota
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kanako C. Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Tomomi Suzuki
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tsuyoshi Shimamura
- Division of Organ Transplantation, Hokkaido University Hospital, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Transplant Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
6
|
Dwyer KM. Burnstock oration - purinergic signalling in kidney transplantation. Purinergic Signal 2022; 18:387-393. [PMID: 35471483 PMCID: PMC9832191 DOI: 10.1007/s11302-022-09865-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/06/2022] [Indexed: 01/14/2023] Open
Abstract
Kidney transplantation is the preferred treatment for individuals with kidney failure offering improved quality and quantity of life. Despite significant advancements in short term graft survival, longer term survival rates have not improved greatly mediated in large by chronic antibody mediated rejection. Strategies to reduce the donor kidney antigenic load may translate to improved transplant survival. CD39 on the vascular endothelium and on circulating cells, in particular regulatory T cells (Treg), is upregulated in response to hypoxic stimuli and plays a critical role in regulating the immune response removing proinflammatory ATP and generating anti-inflammatory adenosine. Herein, the role of CD39 in reducing ischaemia-reperfusion injury (IRI) and on Treg within the context of kidney transplantation is reviewed.
Collapse
Affiliation(s)
- Karen M. Dwyer
- grid.1021.20000 0001 0526 7079School of Medicine, Deakin University, Geelong, 3220 Australia
| |
Collapse
|
7
|
Görner S, Heim C, Weigmann B, von Silva-Tarouca B, Kuckhahn A, Ramsperger-Gleixner M, Zimmermann R, Weyand M, Ensminger SM. Direct Impact of Human Platelets on the Development of Transplant Arteriosclerosis. Transplantation 2022; 106:1180-1192. [PMID: 34468430 DOI: 10.1097/tp.0000000000003935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Platelets play an important role in the pathogenesis of inflammatory and proliferative vascular changes. The aim of this study was to investigate whether human platelets are able to induce transplant arteriosclerosis in a humanized C57/Bl6-Rag2-/-γc-/- mouse xenograft model. METHODS Nonactivated and in vitro-activated human platelets were analyzed and phenotyped for surface markers by flow cytometry. Side branches of human mammary arteries were implanted into the infrarenal aorta of recipients, followed by daily application of human platelets and histological analyzed on day 30 after transplantation. RESULTS Human platelets collected by apheresis had low levels of platelet activation markers. However, after in vitro activation, expression was markedly increased. Sixty minutes after injection in recipient mice, nonactivated human platelets become significantly activated. Increased adhesion of platelets to the vascular endothelium was detected by in vivo fluorescence microscopy. After intravenous injection of nonactivated or activated platelets, human xenografts showed pronounced intimal proliferation. Immunohistological analysis showed that the group treated with activated human platelets exhibited significantly increased intragraft protein expression of intracellular adhesion molecule-1 and platelet-derived growth factor receptor beta and smooth muscle cell migration into the neointima. CONCLUSIONS These data demonstrate that an isolated daily application of both in vivo- and in vitro-activated human platelets results in the development of transplant arteriosclerosis in a humanized mouse transplantation model.
Collapse
Affiliation(s)
- Susann Görner
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | | | - Annika Kuckhahn
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | | | - Robert Zimmermann
- Department of Transfusion Medicine, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Stephan M Ensminger
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
- Present address: Department of Cardiac and Thoracic Vascular Surgery, University Heart Center Lübeck, University Hospital Schleswig Holstein, Lübeck, Germany
| |
Collapse
|
8
|
Goto R, Ito M, Kawamura N, Watanabe M, Ganchiku Y, Kamiyama T, Shimamura T, Taketomi A. The impact of preformed donor‐specific antibodies in living donor liver transplantation according to graft volume. Immun Inflamm Dis 2022; 10:e586. [PMID: 35064772 PMCID: PMC8926496 DOI: 10.1002/iid3.586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/09/2021] [Accepted: 12/31/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Ryoichi Goto
- Department of Gastroenterological Surgery I Hokkaido University Graduate School of Medicine Sapporo Japan
| | - Makoto Ito
- Division of Laboratory and Transfusion Medicine Hokkaido University Hospital Sapporo Japan
| | - Norio Kawamura
- Department of Transplant Surgery Hokkaido University Graduate School of Medicine Sapporo Japan
| | - Masaaki Watanabe
- Department of Transplant Surgery Hokkaido University Graduate School of Medicine Sapporo Japan
| | - Yoshikazu Ganchiku
- Department of Gastroenterological Surgery I Hokkaido University Graduate School of Medicine Sapporo Japan
| | - Toshiya Kamiyama
- Department of Gastroenterological Surgery I Hokkaido University Graduate School of Medicine Sapporo Japan
| | - Tsuyoshi Shimamura
- Division of Organ Transplantation Hokkaido University Hospital Sapporo Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I Hokkaido University Graduate School of Medicine Sapporo Japan
| |
Collapse
|
9
|
Cioni M, Comoli P, Tagliamacco A, Innocente A, Basso S, Fontana I, Magnasco A, Trivelli A, Nocco A, Macchiagodena M, Catenacci L, Klersy C, Verrina E, Garibotto G, Ghiggeri GM, Cardillo M, Ginevri F, Nocera A. Post-transplant de novo non donor-specific HLA antibodies are not associated with poor graft outcome in non-sensitized pediatric recipients of kidney transplantation. Transpl Immunol 2021; 65:101375. [PMID: 33610675 DOI: 10.1016/j.trim.2021.101375] [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: 11/27/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Abstract
While de novo donor-specific HLA antibodies (dnDSAs) have a detrimental impact on kidney graft outcome, the clinical significance of de novo non donor-specific antibodies (dnNDSAs) is more controversial. We retrospectively evaluated for Ab development and characteristics of dnNDSAs serially collected post-transplant sera and, when available, graft biopsy eluates, from 144 non-sensitized, primary pediatric kidney recipients, consecutively transplanted at a single center between 2003 and 2017, using HLA class I and class II single-antigen flow-bead assays (SAB). The results were compared with clinical-pathologic data from HLA antibody negative and HLA dnDSA-positive patients. Forty-five out of 144 patients developed dnNDSAs (31%). Among the dnNDSA-positive patients, 86% displayed one or more class I/II antibodies recognizing antigens included in the CREG/shared epitope groups that also comprise the mismatched donor HLA antigens. Despite potential pathogenicity, as suggested by their occasional presence within the graft, dnNDSAs displayed significantly lower MFI, and limited complement binding and graft homing properties, when compared with dnDSAs. In parallel, the graft survival probability was significantly lower in patients with dnDSA than in those with dnNDSA or without HLA antibodies (p < 0.005). Indeed, the dnNDSA-positive patients remaining dnDSA-negative throughout the posttransplant period did not develop clinical antibody mediated rejection and graft loss, and maintained good graft function at a median follow-up of 9 years. The biological characteristics of dnNDSAs may account for the low graft damaging capability when compared to dnDSAs.
Collapse
Affiliation(s)
- Michela Cioni
- Laboratory of Molecular Nephrology, IRCCS Giannina Gaslini, Genova, Italy
| | - Patrizia Comoli
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Augusto Tagliamacco
- Clinical Nephrology Unit and Transplant Coordination Unit, Policlinico San Martino, Genova, Italy
| | - Annalisa Innocente
- Transplantation Immunology, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Sabrina Basso
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Iris Fontana
- Vascular and Endovascular Unit and Kidney Transplant Surgery Unit, Ospedale Policlinico San Martino, Genova, Italy
| | - Alberto Magnasco
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy
| | - Antonella Trivelli
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy
| | - Angela Nocco
- Transplantation Immunology, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Macchiagodena
- Transplantation Immunology, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Laura Catenacci
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Catherine Klersy
- Biometry and Clinical Epidemiology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Enrico Verrina
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy
| | - Giacomo Garibotto
- Clinical Nephrology Unit, University of Genova and Policlinico San Martino Genova, Italy
| | - Gian Marco Ghiggeri
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy
| | - Massimo Cardillo
- Transplantation Immunology, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy; Italian National Transplant Centre, Italian National Institute of Health (ISS), Rome, Italy
| | - Fabrizio Ginevri
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy.
| | - Arcangelo Nocera
- Nephrology, Dialysis, Transplantation Unit, IRCCS Giannina Gaslini, Genova, Italy
| |
Collapse
|
10
|
Zheng H, Su Y, Zhu C, Quan D, Skaro AI, McAlister V, Lacefield JC, Jiang J, Xue P, Wang Y, Zheng X. An Addition of U0126 Protecting Heart Grafts From Prolonged Cold Ischemia-Reperfusion Injury in Heart Transplantation: A New Preservation Strategy. Transplantation 2021; 105:308-317. [PMID: 32776778 DOI: 10.1097/tp.0000000000003402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is the major cause of primary graft dysfunction in organ transplantation. The mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) signaling pathway plays a crucial role in cell physiological and pathological processes including IRI. This study aims to investigate whether inhibition of ERK signaling with U0126 can prevent prolonged cold IRI in heart transplantation. METHODS Rat cardiac cell line H9c2 cells were treated with U0126 before exposure to hypothermic hypoxia/reoxygenation (H/R) conditions. The effect of U0126 on H9c2 cells in response to H/R stress was determined by measuring cell death, reactive oxygen species production, mitochondrial membrane potential, and ERK signaling activation. Mouse syngeneic heterotopic heart transplantation was conducted, where a donor heart was preserved in the University of Wisconsin (UW) solution supplemented with U0126 for 24 hours at 4°C before transplantation. Heart graft function, histopathologic changes, apoptosis, and fibrosis were measured to assess IRI. RESULTS Phosphorylated ERK was increased in both in vitro H/R-injured H9c2 cells and in vivo heart grafts with IRI. Pretreatment with U0126 inhibited ERK phosphorylation and prevented H9c2 cells from cell death, reactive oxygen species generation, and mitochondrial membrane potential loss in response to H/R. Preservation of donor hearts with U0126-supplemented solution improved graft function and reduced IRI by reductions in cell apoptosis/death, neutrophil infiltration, and fibrosis of the graft. CONCLUSIONS Addition of U0126 to UW solution reduces ERK signal activation and attenuates prolonged cold IRI in a heart transplantation model. ERK inhibition with U0126 may be a useful strategy to minimize IRI in organ transplantation.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- College of Life Science, Wuhan University, Wuhan, China
| | - Yale Su
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Douglas Quan
- Department of Surgery, Western University, London, ON, Canada
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
| | - Anton I Skaro
- Department of Surgery, Western University, London, ON, Canada
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
| | - Vivian McAlister
- Department of Surgery, Western University, London, ON, Canada
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
| | - James C Lacefield
- Department of Electrical and Computer Engineering, Western University, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
- Department of Oncology, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Jifu Jiang
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
| | - Peng Xue
- College of Life Science, Wuhan University, Wuhan, China
| | - Yefu Wang
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Xiufen Zheng
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Department of Surgery, Western University, London, ON, Canada
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
- Department of Oncology, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
11
|
Uehara M, Solhjou Z, Banouni N, Kasinath V, Xiaqun Y, Dai L, Yilmam O, Yilmaz M, Ichimura T, Fiorina P, Martins PN, Ohori S, Guleria I, Maarouf OH, Tullius SG, McGrath MM, Abdi R. Ischemia augments alloimmune injury through IL-6-driven CD4 + alloreactivity. Sci Rep 2018; 8:2461. [PMID: 29410442 PMCID: PMC5802749 DOI: 10.1038/s41598-018-20858-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023] Open
Abstract
Ischemia reperfusion injuries (IRI) are unavoidable in solid organ transplantation. IRI augments alloimmunity but the mechanisms involved are poorly understood. Herein, we examined the effect of IRI on antigen specific alloimmunity. We demonstrate that ischemia promotes alloimmune activation, leading to more severe histological features of rejection, and increased CD4+ and CD8+ T cell graft infiltration, with a predominantly CD8+ IFNγ+ infiltrate. This process is dependent on the presence of alloreactive CD4+ T cells, where depletion prevented infiltration of ischemic grafts by CD8+ IFNγ+ T cells. IL-6 is a known driver of ischemia-induced rejection. Herein, depletion of donor antigen-presenting cells reduced ischemia-induced CD8+ IFNγ+ allograft infiltration, and improved allograft outcomes. Following prolonged ischemia, accelerated rejection was observed despite treatment with CTLA4Ig, indicating that T cell costimulatory blockade failed to overcome the immune activating effect of IRI. However, despite severe ischemic injury, treatment with anti-IL-6 and CTLA4Ig blocked IRI-induced alloimmune injury and markedly improved allograft survival. We describe a novel pathway where IRI activates innate immunity, leading to upregulation of antigen specific alloimmunity, resulting in chronic allograft injury. Based on these findings, we describe a clinically relevant treatment strategy to overcome the deleterious effect of IRI, and provide superior long-term allograft outcomes.
Collapse
Affiliation(s)
- Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ye Xiaqun
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Dai
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Osman Yilmam
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mine Yilmaz
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Fiorina
- Division of Nephrology, Boston Children Hospital, Harvard Medical School, Boston, MA, USA
| | - Paulo N Martins
- Division of Surgery, University of Massachusetts Medical School, Boston, MA, USA
| | - Shunsuke Ohori
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Indira Guleria
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar H Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martina M McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Cherif-Sayadi A, Hadj Ayed-Tka K, Zaouali MA, Bejaoui M, Hadj-Abdallah N, Bouhlel A, Ben Abdennebi H. Nitrite enhances liver graft protection against cold ischemia reperfusion injury through a NOS independent pathway. Libyan J Med 2017; 12:1308780. [PMID: 28357909 PMCID: PMC5418943 DOI: 10.1080/19932820.2017.1308780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/12/2017] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION Nitrite has been found to protect liver graft from cold preservation injury. However, the cell signaling pathway involved in this protection remains unclear. Here, we attempt to clarify if the NOS pathway by using the NOS inhibitor, L-NAME (L-NG-Nitroarginine methyl ester). ANIMALS AND METHODS Rat livers were conserved for 24 h at 4°C in (IGL-1) solution enriched or not with nitrite at 50 nM. In a third group, rats were pretreated with 50 mg/kg of L-NAME before their liver procurement and preservation in IGL-1 supplemented with nitrite (50 nM) and L-NAME (1 mM). After 24 h of cold storage, rat livers were ex-vivo perfused at 37°C during 2 h. Control livers were perfused without cold storage. RESULTS Nitrite effectively protected the rat liver grafts from the onset of cold I/R injury. L-NAME treatment did not abolish the beneficial effects of nitrite. Liver damage, protein oxidation and lipid peroxidation remained at low levels in both nitrite-treated groups when compared to IGL-1 group. Antioxidant enzyme activities and functional parameters were unchanged after NOS inhibition. CONCLUSION Despite NOS inhibition by L-NAME, nitrite can still provide hepatic protection during cold I/R preservation. This suggests that nitrite acts through a NOS-independent pathway.
Collapse
Affiliation(s)
- Amani Cherif-Sayadi
- Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Kaouther Hadj Ayed-Tka
- Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Mohamed Amine Zaouali
- Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
- High Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Mohamed Bejaoui
- Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Najet Hadj-Abdallah
- High Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Ahlem Bouhlel
- High Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Hassen Ben Abdennebi
- Research Unit of Biology and Molecular Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
13
|
Green ML, Breite AG, Beechler CA, Dwulet FE, McCarthy RC. Effectiveness of different molecular forms of C. histolyticum class I collagenase to recover islets. Islets 2017; 9:177-181. [PMID: 28933628 PMCID: PMC5710699 DOI: 10.1080/19382014.2017.1365996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
One factor that may contribute to variability between different lots of purified collagenase to recover islets is the molecular form of C. histolyticum class I (C1) collagenase used in the isolation procedure. Two different enzyme mixtures containing C1, class II (C2) collagenase and BP Protease were compared for their effectiveness to recover islets from split adult porcine pancreas. The same enzyme activities per g trimmed tissue were used for all isolations with the only difference being the mass of C1 required to achieve 25,000 collagen degradation activity U/g tissue. The results show no differences in performance of the two enzyme mixtures. The only significant difference is 19 fold more truncated C1 was required to achieve the same result as intact C1.
Collapse
Affiliation(s)
| | | | | | | | - Robert C. McCarthy
- VitaCyte LLC, Indianapolis, IN, USA
- CONTACT Robert C. McCarthy VitaCyte LLC, 1341 Sadlier Circle West Drive, Indianapolis, IN 46239, USA
| |
Collapse
|
14
|
Siemeni T, Knöfel AK, Madrahimov N, Sommer W, Avsar M, Salman J, Ius F, Frank N, Büchler G, Jonigk D, Jansson K, Maus U, Tudorache I, Falk CS, Haverich A, Warnecke G. In Vivo Development of Transplant Arteriosclerosis in Humanized Mice Reflects Alloantigen Recognition and Peripheral Treg Phenotype of Lung Transplant Recipients. Am J Transplant 2016; 16:3150-3162. [PMID: 27273729 DOI: 10.1111/ajt.13905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Experimentally, regulatory T cells inhibit rejection. In clinical transplantations, however, it is not known whether T cell regulation is the cause for, or an epiphenomenon of, long-term allograft survival. Here, we study naïve and alloantigen-primed T cell responses of clinical lung transplant recipients in humanized mice. The pericardiophrenic artery procured from human lung grafts was implanted into the aorta of NODrag-/- /IL-2rγc-/- mice reconstituted with peripheral blood mononuclear cells (PBMCs) from the respective lung recipient. Naïve or primed allogeneic PBMCs procured 21 days post-lung transplantation with or without enriching for CD4+ CD25high T cells were used. Transplant arteriosclerosis was assessed 28 days later by histology. Mice reconstituted with alloantigen-primed PBMCs showed significantly more severe transplant arteriosclerosis than did mice with naïve PBMCs (p = 0.005). Transplant arteriosclerosis was equally suppressed by enriching for autologous naïve (p = 0.012) or alloantigen-primed regulatory T cells (Tregs) (p = 0.009). Alloantigen priming in clinical lung recipients can be adoptively transferred into a humanized mouse model. Transplant arteriosclerosis elicited by naïve or alloantigen-primed PBMCs can be similarly controlled by potent autologous Tregs. Cellular therapy with expanded autologous Tregs in lung transplantation might be a promising future strategy.
Collapse
Affiliation(s)
- T Siemeni
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - A-K Knöfel
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - N Madrahimov
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - W Sommer
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - M Avsar
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - J Salman
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - F Ius
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - N Frank
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - G Büchler
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - D Jonigk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - K Jansson
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - U Maus
- German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.,Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - I Tudorache
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - C S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - G Warnecke
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany. .,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
15
|
Kleinert E, Langenmayer MC, Reichart B, Kindermann J, Griemert B, Blutke A, Troidl K, Mayr T, Grantzow T, Noyan F, Abicht JM, Fischer S, Preissner KT, Wanke R, Deindl E, Guethoff S. Ribonuclease (RNase) Prolongs Survival of Grafts in Experimental Heart Transplantation. J Am Heart Assoc 2016; 5:e003429. [PMID: 27121849 PMCID: PMC4889206 DOI: 10.1161/jaha.116.003429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 03/03/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cell damage, tissue and vascular injury are associated with the exposure and release of intracellular components such as RNA, which promote inflammatory reactions and thrombosis. Based on the counteracting anti-inflammatory and cardioprotective functions of ribonuclease A (RNase A) in this context, its role in an experimental model of heart transplantation in rats was studied. METHODS AND RESULTS Inbred BN/OrlRj rat cardiac allografts were heterotopically transplanted into inbred LEW/OrlRj rats. Recipients were intravenously treated every other day with saline or bovine pancreatic RNase A (50 μg/kg). Toxic side effects were not found (macroscopically and histologically). Heart tissue flow cytometry and quantitative morphological analyses of explanted hearts at postoperative day 1 or postoperative day 4 showed reduced leukocyte infiltration, edema, and thrombus formation in RNase A-treated rats. In allogeneic mixed lymphocyte reactions, RNase A decreased the proliferation of effector T cells. RNase A treatment of rats resulted in prolonged median graft survival up to 10.5 days (interquartile range 1.8) compared to 6.5 days (interquartile range 1.0) in saline treatment (P=0.001). Treatment of rats with a new generated (recombinant) human pancreatic RNase 1 prolonged median graft survival similarly, unlike treatment with (recombinant) inactive human RNase 1 (each 50 μg/kg IV every other day, 11.0 days, interquartile range 0.3, versus 8.0 days, interquartile range 0.5, P=0.007). CONCLUSIONS Upon heart transplantation, RNase administration appears to present a promising and safe drug to counteract ischemia/reperfusion injury and graft rejection. Furthermore, RNase treatment may be considered in situations of critical reperfusion after percutaneous coronary interventions or in cardiac surgery using the heart-lung machine.
Collapse
Affiliation(s)
- Eike Kleinert
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Martin C Langenmayer
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, Germany
| | - Bruno Reichart
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Jana Kindermann
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Germany
| | - Barbara Griemert
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Kerstin Troidl
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany Department of Vascular and Endovascular Surgery, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tanja Mayr
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Anaesthesiology, Ludwig-Maximilians-Universität München, Germany
| | - Tobias Grantzow
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jan-Michael Abicht
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Anaesthesiology, Ludwig-Maximilians-Universität München, Germany
| | - Silvia Fischer
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Ruediger Wanke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Elisabeth Deindl
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Sonja Guethoff
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
16
|
Kenney LL, Shultz LD, Greiner DL, Brehm MA. Humanized Mouse Models for Transplant Immunology. Am J Transplant 2016; 16:389-97. [PMID: 26588186 PMCID: PMC5283075 DOI: 10.1111/ajt.13520] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/25/2023]
Abstract
Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.
Collapse
Affiliation(s)
- Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| | | | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605,Corresponding Author: Dale L. Greiner, PhD, University of Massachusetts Medical School, 368 Plantation Street, AS7-2051, Worcester, MA 01605, Office: 508-856-1911, Fax: 508-856-4093,
| | - Michael A. Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| |
Collapse
|
17
|
Absence of Activation-induced Cytidine Deaminase, a Regulator of Class Switch Recombination and Hypermutation in B Cells, Suppresses Aorta Allograft Vasculopathy in Mice. Transplantation 2015; 99:1598-605. [PMID: 25769064 DOI: 10.1097/tp.0000000000000688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antibody-mediated rejection is caused in part by increasing circulation/production of donor-specific antibody (DSA). Activation-induced cytidine deaminase (AID) is a key regulator of class switch recombination and somatic hypermutation of immunoglobulin in B cells, yet its role in antibody-mediated transplant rejection remains unclear. We show here that AID deficiency in mice enables suppression of allograft vasculopathy (AV) after aorta transplantation, a DSA-mediated process. METHODS Splenocytes from C57BL/6 J (B6) AID(−/−) mice were used for determining in vitro proliferation responses, alloreactivity, cell surface marker expression, and antibody production. BALB/c mouse aortas were transplanted into B6 AID(−/−) mice with or without FK506 treatment. Blood and aorta grafts were harvested on day 30 after transplantation and were subjected to DSA, histological, and immunohistological analyses. RESULTS The AID(−/−) splenocytes were comparable to wild type splenocytes in proliferation responses, alloreactivity, and expression of cell surface markers in vitro. However, they completely failed to produce immunoglobulin G, although they were not impaired in immunoglobulin M production relative to controls. Furthermore, BALB/c aorta grafts from B6 AID(−/−) recipient mice on day 30 after transplantation showed reduced signs of AV compared to the grafts from B6 wild type recipient mice which had severe vascular intimal hyperplasia, interstitial fibrosis, and inflammation. Treatment with FK506 produced a synergistic effect in the grafts from AID(−/−) recipients with further reduction of intimal hyperplasia and fibrosis scores. CONCLUSIONS The AID deficiency inhibits DSA-mediated AV after aorta transplantation in mice. We propose that AID could be a novel molecular target for controlling antibody-mediated rejection in organ transplantation.
Collapse
|
18
|
Süsal C, Wettstein D, Döhler B, Morath C, Ruhenstroth A, Scherer S, Tran TH, Gombos P, Schemmer P, Wagner E, Fehr T, Živčić-Ćosić S, Balen S, Weimer R, Slavcev A, Bösmüller C, Norman DJ, Zeier M, Opelz G. Association of Kidney Graft Loss With De Novo Produced Donor-Specific and Non-Donor-Specific HLA Antibodies Detected by Single Antigen Testing. Transplantation 2015; 99:1976-1980. [PMID: 25769065 DOI: 10.1097/tp.0000000000000672] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The association of donor-specific HLA antibodies (DSA) with kidney graft failure has been addressed previously; however, the majority of studies were based on small numbers of patients with graft failure. METHODS We investigated 83 patients with failed kidney transplants for a possible association of de novo development and persistence or loss of pre-existing DSA with graft failure. Single Antigen Bead assay-detected DSA and non-DSA antibodies were compared between patients with graft loss and matched controls with functioning grafts. RESULTS The incidence of weak de novo DSA or non-DSA at a mean fluorescence intensity of 500 or higher was higher in the graft loss than in the nonrejector group (76% vs 40%, P < 0.001). Because of the low number of patients developing de novo DSA, the DSA results did not reach statistical significance (only 22% of patients with graft loss developed de novo DSA). However, at all cutoffs, there was a significantly higher rate of graft loss in patients with de novo non-DSA. The incidence of strong pretransplant DSA that persist after transplantation was higher in the graft loss group (10% vs 1%, P = 0.034). When C1q-binding ability in sera of rejectors and nonrejectors with posttransplant de novo or persistent DSA was compared, none of the nonrejectors demonstrated C1q positivity, whereas 43% of patients with graft loss showed C1q-positive antibodies, although not necessarily donor-specific (P < 0.001). CONCLUSIONS Our data show that the posttransplant presence of persisting or de novo HLA antibodies, especially if C1q binding, is associated with graft loss, even if the antibodies are not specific for mismatched donor HLA.
Collapse
Affiliation(s)
- Caner Süsal
- 1 Department of Transplantation Immunology, University of Heidelberg, Heidelberg, Germany. 2 Department of Nephrology, University of Heidelberg, Heidelberg, Germany. 3 Department of Transplantation and General Surgery, University of Heidelberg, Heidelberg, Germany. 4 Immunology and Histocompatibility Laboratory, CHU de Quebec, Quebec, QC, Canada. 5 Department of Microbiology, Infectious Diseases and Immunology, Laval University, Quebec, QC, Canada. 6 Division of Nephrology, University Hospital Zurich, Switzerland. 7 Departments of Nephrology, Dialysis, Kidney Transplantation and Transfusion Medicine, University Hospital Centre, Rijeka, Croatia. 8 Department of Internal Medicine, University Clinic of Giessen and Marburg, Campus Giessen, Giessen, Germany. 9 Department of Immunogenetics, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic. 10 Department of General and Transplant Surgery, Innsbruck Medical University, Innsbruck, Austria. 11 Oregon Health and Science University, Portland, OR
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mori DN, Kreisel D, Fullerton JN, Gilroy DW, Goldstein DR. Inflammatory triggers of acute rejection of organ allografts. Immunol Rev 2015; 258:132-44. [PMID: 24517430 DOI: 10.1111/imr.12146] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Solid organ transplantation is a vital therapy for end stage diseases. Decades of research have established that components of the adaptive immune system are critical for transplant rejection, but the role of the innate immune system in organ transplantation is just emerging. Accumulating evidence indicates that the innate immune system is activated at the time of organ implantation by the release of endogenous inflammatory triggers. This review discusses the nature of these triggers in organ transplantation and also potential mediators that may enhance inflammation resolution after organ implantation.
Collapse
Affiliation(s)
- Daniel N Mori
- Departments of Internal Medicine and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | | |
Collapse
|
20
|
Khader A, Yang WL, Kuncewitch M, Prince JM, Marambaud P, Nicastro J, Coppa GF, Wang P. Novel resveratrol analogues attenuate renal ischemic injury in rats. J Surg Res 2015; 193:807-15. [PMID: 25214260 PMCID: PMC4268227 DOI: 10.1016/j.jss.2014.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/20/2014] [Accepted: 08/08/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion (I/R) is a severe clinical complication with no specific treatment. Resveratrol has been shown as a promising experimental agent in renal I/R due to its effect on cellular energy metabolism, oxidative stress, and inflammation. Recently, we identified two biologically active resveratrol analogues (RSVAs), RSVA405 and RSVA314. We hypothesized that both RSAVs would attenuate I/R-induced renal injury. METHODS Adult male rats were subjected to renal I/R through bilateral renal pedicle clamping for 60 min, followed by reperfusion. RSVA405 (3 mg/kg Body Weight), RSVA314 (3 mg/kg Body Weight), or vehicle (10% dimethyl sulfoxide and 33% Solutol in phosphate buffered saline) were administered by intraperitoneal injection 1 h before ischemia. Blood and renal tissues were collected 24 h after I/R for evaluation. RESULTS Administration of RSVA405 and RSVA314 significantly reduced the serum levels of renal dysfunction and injury markers, including creatinine, blood urea nitrogen, aspartate aminotransferase, and lactate dehydrogenase, compared with vehicle. The protective effect of RSVA405 and RSVA314 was also reflected on histologic evaluation. Both RSVAs reduced the number of apoptotic cells by more than 60% as determined by transferase dUTP nick end labeling assay, compared with vehicle. The renal adenosine triphosphate levels of the vehicle group was decreased to 52.4% of control, whereas those of the RSVA405 and RSVA314 groups were restored to 72.3% and 79.6% of control, respectively. Both RSVAs significantly reduced the protein expression of inducible nitric oxide synthase and nitrotyrosine and the messenger RNA levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β. CONCLUSIONS RSVA405 and RSVA314 attenuate I/R-induced renal injury through the modulation of energy metabolism, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Adam Khader
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Weng-Lang Yang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Michael Kuncewitch
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| | - Jose M Prince
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Jeffrey Nicastro
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| | - Gene F Coppa
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| | - Ping Wang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York; Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, New York.
| |
Collapse
|
21
|
Abe T, Su CA, Iida S, Baldwin WM, Nonomura N, Takahara S, Fairchild RL. Graft-derived CCL2 increases graft injury during antibody-mediated rejection of cardiac allografts. Am J Transplant 2014; 14:1753-64. [PMID: 25040187 PMCID: PMC4464804 DOI: 10.1111/ajt.12780] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/14/2014] [Accepted: 04/01/2014] [Indexed: 01/25/2023]
Abstract
The pathogenic role of macrophages in antibody-mediated rejection (AMR) remains unclear. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is a potent chemotactic factor for monocytes and macrophages. The current studies used a murine model of AMR to investigate the role of graft-derived CCL2 in AMR and how macrophages may participate in antibody-mediated allograft injury. B6.CCR5−/−/CD8−/− recipients rejected MHC-mismatched WT A/J allografts with high donor-reactive antibody titers and diffuse C4d deposition in the large vessels and myocardial capillaries, features consistent with AMR. In contrast, A/J.CCL2−/− allografts induced low donor-reactive antibody titers and C4d deposition at Day 7 posttransplant. Decreased donor-reactive CD4 T cells producing interferon gamma were induced in response to A/J.CCL2−/− versus WT allografts. Consequently, A/J.CCL2−/− allograft survival was modestly but significantly longer than A/J allografts. Macrophages purified from WT allografts expressed high levels of IL-1β and IL-12p40 and this expression and the numbers of classically activated macrophages were markedly reduced in CCL2-deficient allografts on Day 7. The results indicate that allograft-derived CCL2 plays an important role in directing classically activated macrophages into allografts during AMR and that macrophages are important contributors to the inflammatory environment mediating graft tissue injury in this pathology, suggesting CCL2 as a therapeutic target for AMR.
Collapse
Affiliation(s)
- Toyofumi Abe
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Charles A. Su
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Shoichi Iida
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - William M. Baldwin
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Norio Nonomura
- Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shiro Takahara
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Robert L. Fairchild
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
22
|
Sirtuin 1 Activation Stimulates Mitochondrial Biogenesis and Attenuates Renal Injury After Ischemia-Reperfusion. Transplantation 2014; 98:148-56. [DOI: 10.1097/tp.0000000000000194] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|