1
|
Bery AI, Belousova N, Hachem RR, Roux A, Kreisel D. Chronic Lung Allograft Dysfunction: Clinical Manifestations and Immunologic Mechanisms. Transplantation 2025; 109:454-466. [PMID: 39104003 PMCID: PMC11799353 DOI: 10.1097/tp.0000000000005162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
The term "chronic lung allograft dysfunction" has emerged to describe the clinical syndrome of progressive, largely irreversible dysfunction of pulmonary allografts. This umbrella term comprises 2 major clinical phenotypes: bronchiolitis obliterans syndrome and restrictive allograft syndrome. Here, we discuss the clinical manifestations, diagnostic challenges, and potential therapeutic avenues to address this major barrier to improved long-term outcomes. In addition, we review the immunologic mechanisms thought to propagate each phenotype of chronic lung allograft dysfunction, discuss the various models used to study this process, describe potential therapeutic targets, and identify key unknowns that must be evaluated by future research strategies.
Collapse
Affiliation(s)
- Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Natalia Belousova
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Ramsey R Hachem
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
- Paris Transplant Group, INSERM U 970s, Paris, France
| | - Daniel Kreisel
- Departments of Surgery, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
2
|
Kang M, Park HK, Kim KS, Choi D. Animal models for transplant immunology: bridging bench to bedside. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:354-376. [PMID: 39233453 PMCID: PMC11732767 DOI: 10.4285/ctr.24.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 09/06/2024]
Abstract
The progress of transplantation has been propelled forward by animal experiments. Animal models have not only provided opportunities to understand complex immune mechanisms in transplantation but also served as a platform to assess therapeutic interventions. While small animals have been instrumental in uncovering new therapeutic concepts related to immunosuppression and immune tolerance, the progression to human trials has largely been driven by studies in large animals. Recent research has begun to explore the potential of porcine organs to address the shortage of available organs. The consistent progress in transplant immunology research can be attributed to a thorough understanding of animal models. This review provides a comprehensive overview of the available animal models, detailing their modifications, strengths, and weaknesses, as well as their historical applications, to aid researchers in selecting the most suitable model for their specific research needs.
Collapse
Affiliation(s)
- Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Hwon Kyum Park
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Kyeong Sik Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
3
|
Figueiredo C, Chen-Wacker C, Salman J, Carvalho-Oliveira M, Monthé TS, Höffler K, Rother T, Hacker K, Valdivia E, Pogozhykh O, Hammer S, Sommer W, Yuzefovych Y, Wenzel N, Haverich A, Warnecke G, Blasczyk R. Knockdown of swine leukocyte antigen expression in porcine lung transplants enables graft survival without immunosuppression. Sci Transl Med 2024; 16:eadi9548. [PMID: 39018368 DOI: 10.1126/scitranslmed.adi9548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 12/21/2023] [Accepted: 06/21/2024] [Indexed: 07/19/2024]
Abstract
Immune rejection remains the major obstacle to long-term survival of allogeneic lung transplants. The expression of major histocompatibility complex molecules and minor histocompatibility antigens triggers allogeneic immune responses that can lead to allograft rejection. Transplant outcomes therefore depend on long-term immunosuppression, which is associated with severe side effects. To address this problem, we investigated the effect of genetically engineered transplants with permanently down-regulated swine leukocyte antigen (SLA) expression to prevent rejection in a porcine allogeneic lung transplantation (LTx) model. Minipig donor lungs with unmodified SLA expression (control group, n = 7) or with modified SLA expression (treatment group, n = 7) were used to evaluate the effects of SLA knockdown on allograft survival and on the nature and strength of immune responses after terminating an initial 4-week period of immunosuppression after LTx. Genetic engineering to down-regulate SLA expression was achieved during ex vivo lung perfusion by lentiviral transduction of short hairpin RNAs targeting mRNAs encoding β2-microglobulin and class II transactivator. Whereas all grafts in the control group were rejected within 3 months, five of seven animals in the treatment group maintained graft survival without immunosuppression during the 2-year monitoring period. Compared with controls, SLA-silenced lung recipients had lower donor-specific antibodies and proinflammatory cytokine concentrations in the serum. Together, these data demonstrate a survival benefit of SLA-down-regulated lung transplants in the absence of immunosuppression.
Collapse
Affiliation(s)
- Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Chen Chen-Wacker
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Jawad Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Marco Carvalho-Oliveira
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | | | - Klaus Höffler
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Tamina Rother
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Karolin Hacker
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Emilio Valdivia
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Olena Pogozhykh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Hammer
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Wiebke Sommer
- Department of Cardiac Surgery, University of Kiel, 24105 Kiel, Germany
| | - Yuliia Yuzefovych
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Nadine Wenzel
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Gregor Warnecke
- Department of Cardiac Surgery, University of Kiel, 24105 Kiel, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
4
|
Pai AC, Parekh KR, Engelhardt JF, Lynch TJ. Ferret respiratory disease models for the study of lung stem cells. LUNG STEM CELLS IN DEVELOPMENT, HEALTH AND DISEASE 2021:273-289. [DOI: 10.1183/2312508x.10010320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
5
|
Kitano K, Ohata K, Economopoulos KP, Gorman DE, Gilpin SE, Becerra DC, Ott HC. Orthotopic Transplantation of Human Bioartificial Lung Grafts in a Porcine Model: A Feasibility Study. Semin Thorac Cardiovasc Surg 2021; 34:752-759. [PMID: 33713829 DOI: 10.1053/j.semtcvs.2021.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/04/2021] [Indexed: 12/31/2022]
Abstract
Lung transplantation is the only treatment for end-stage lung disease; however, donor organ shortage and intense immunosuppression limit its broad clinical impact. Bioengineering of lungs with patient-derived cells could overcome these problems. We created bioartificial lungs by seeding human-derived cells onto porcine lung matrices and performed orthotopic transplantation to assess feasibility and in vivo function. Porcine decellularized lung scaffolds were seeded with human airway epithelial cells and human umbilical vein endothelial cells. Following in vitro culture, the bioartificial lungs were orthotopically transplanted into porcine recipients with planned 1-day survival (n = 3). Lungs were assessed with histology and in vivo function. Orthotopic transplantation of cadaveric lungs was performed as control. Engraftment of endothelial and epithelial cells in the grafts were histologically demonstrated. Technically successful orthotopic anastomoses of the vasculatures and airway were achieved in all animals. Perfusion and ventilation of the lung grafts were confirmed intraoperatively. The gas exchange function was evident immediately after transplantation; PO2 gradient between pulmonary artery and vein were 178 ± 153 mm Hg in the bioartificial lung group and 183 ± 117 mm Hg in the control group. At time of evaluation 24 hours after reperfusion, the pulmonary arteries were found to be occluded with thrombus in all bioartificial lungs. Engineering and orthotopic transplantation of bioartificial lungs with human cells were technically feasible in a porcine model. Early gas exchange function was evident. Further progress in optimizing recellularization and maturation of the grafts will be necessary for sustained perfusability and function.
Collapse
Affiliation(s)
- Kentaro Kitano
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Thoracic Surgery, The University of Tokyo Hospital, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Keiji Ohata
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Daniel E Gorman
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sarah E Gilpin
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David C Becerra
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Harald C Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
6
|
Why some organ allografts are tolerated better than others: new insights for an old question. Curr Opin Organ Transplant 2020; 24:49-57. [PMID: 30516578 DOI: 10.1097/mot.0000000000000594] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW There is great variability in how different organ allografts respond to the same tolerance induction protocol. Well known examples of this phenomenon include the protolerogenic nature of kidney and liver allografts as opposed to the tolerance-resistance of heart and lung allografts. This suggests there are organ-specific factors which differentially drive the immune response following transplantation. RECENT FINDINGS The specific cells or cell products that make one organ allograft more likely to be accepted off immunosuppression than another are largely unknown. However, new insights have been made in this area recently. SUMMARY The current review will focus on the organ-intrinsic factors that contribute to the organ-specific differences observed in tolerance induction with a view to developing therapeutic strategies to better prevent organ rejection and promote tolerance induction of all organs.
Collapse
|
7
|
Mariscal A, Caldarone L, Tikkanen J, Nakajima D, Chen M, Yeung J, Cypel M, Liu M, Keshavjee S. Pig lung transplant survival model. Nat Protoc 2019; 13:1814-1828. [PMID: 30072720 DOI: 10.1038/s41596-018-0019-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although lung transplant is a life-saving therapy for some patients, primary graft dysfunction (PGD) is a leading cause of mortality and morbidity soon after a transplant. Ischemia reperfusion injury is known to be one of the most critical factors in PGD development. PGD is by definition an acute lung injury syndrome that occurs during the first 3 d following lung transplantation. To successfully translate laboratory discoveries to clinical practice, a reliable and practical large animal model is critical. This protocol describes a surgical technique for swine lung transplantation and postoperative management for a further 3 d post transplant. The protocol includes the background and rationale, required supplies, and a detailed description of the donor operation, transplant surgery, postoperative care, and sacrifice surgery. A pig lung transplant model is reliably produced in which the recipients survive for 3 d post transplant. This 3-d survival model can be used by lung transplant researchers to assess the development of PGD and to test therapeutic strategies targeting PGD. In total, the protocol requires 5 h for the surgeries, plus ~2 h in total for the postoperative care.
Collapse
Affiliation(s)
- Andrea Mariscal
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Lindsay Caldarone
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jussi Tikkanen
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Manyin Chen
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada
| | - Jonathan Yeung
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Marcelo Cypel
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| | - Shaf Keshavjee
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada. .,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Diagnosis, Pathophysiology and Experimental Models of Chronic Lung Allograft Rejection. Transplantation 2019; 102:1459-1466. [PMID: 29683998 DOI: 10.1097/tp.0000000000002250] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chronic rejection is the Achilles heel of modern lung transplantation, characterized by a slow, progressive decline in allograft function. Clinically, this manifests as obstructive disease, restrictive disease, or a mixture of the 2 depending on the underlying pathology. The 2 major phenotypes of chronic rejection include bronchiolitis obliterans syndrome and restrictive allograft syndrome. The last decade of research has revealed that each of these phenotypes has a unique underlying pathophysiology which may require a distinct treatment regimen for optimal control. Insights into the intricate alloimmune pathways contributing to chronic rejection have been gained from both large and small animal models, suggesting directions for future research. In this review, we explore the pathological hallmarks of chronic rejection, recent insights gained from both clinical and basic science research, and the current state of animal models of chronic lung rejection.
Collapse
|
9
|
Salman J, Jansson K, Siemeni T, Sommer W, Knoefel AK, Ahrens L, Nakagiri T, Ius F, Tudorache I, Kruse B, Thissen S, Jonigk D, Strüber M, Haverich A, Warnecke G, Avsar M. Role for primary immunosuppression with everolimus after pulmonary transplantation. Transpl Immunol 2018; 49:12-19. [PMID: 29588160 DOI: 10.1016/j.trim.2018.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 12/31/2022]
Affiliation(s)
- J Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany.
| | - K Jansson
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Th Siemeni
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - W Sommer
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - A-K Knoefel
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - L Ahrens
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - T Nakagiri
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - F Ius
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - I Tudorache
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - B Kruse
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - S Thissen
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - D Jonigk
- Institute for Pathology, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - M Strüber
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - G Warnecke
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - M Avsar
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Gauthier JM, Hachem RR, Kreisel D. Update on Chronic Lung Allograft Dysfunction. CURRENT TRANSPLANTATION REPORTS 2016; 3:185-191. [PMID: 28090432 DOI: 10.1007/s40472-016-0112-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) encompasses a range of pathologies that cause a transplanted lung to not achieve or maintain normal function. CLAD manifests as airflow restriction and/or obstruction and is predominantly a result of chronic rejection. Three distinct phenotypes of chronic rejection are now recognized: bronchiolitis obliterans, neutrophilic reversible allograft dysfunction, and restrictive allograft syndrome. Recent investigations have revealed that each phenotype has a unique pathology and histopathological findings, suggesting that treatment regimens should be tailored to the underlying etiology. CLAD is poorly responsive to treatment once diagnosed, and therefore the prevention of the factors that predispose a patient to develop CLAD is critically important. Small and large animal models have contributed significantly to our understanding of CLAD and more studies are needed to develop treatment regimens that are effective in humans.
Collapse
Affiliation(s)
- Jason M Gauthier
- Department of Surgery, Washington University in St. Louis, Saint Louis, MO
| | - Ramsey R Hachem
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University in St. Louis, Saint Louis, MO ; Department of Pathology & Immunology, Washington University in St. Louis, Saint Louis, MO
| |
Collapse
|
11
|
Recipient-matching of Passenger Leukocytes Prolongs Survival of Donor Lung Allografts in Miniature Swine. Transplantation 2016; 99:1372-8. [PMID: 25757217 DOI: 10.1097/tp.0000000000000676] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Allograft rejection continues to be a vexing problem in clinical lung transplantation, and the role played by passenger leukocytes in the rejection or acceptance of an organ is unclear. We tested whether recipient-matching of donor graft passenger leukocytes would impact graft survival in a preclinical model of orthotopic left lung transplantation. METHODS In the experimental group (group 1), donor lungs were obtained from chimeric swine, in which the passenger leukocytes (but not the parenchyma) were major histocompatibility complex-matched to the recipients (n = 3). In the control group (group 2), both the donor parenchyma and the passenger leukocytes were major histocompatibility complex-mismatched to the recipients (n = 3). RESULTS Lungs harvested from swine previously rendered chimeric by hematopoietic stem cell transplantation using recipient-type cells showed a high degree of passenger leukocyte chimerism by immunohistochemistry and flow cytometry. The chimeric lungs containing passenger leukocytes matched to the lung recipient (group 1) survived on average 107 days (range, 80-156). Control lung allografts (group 2) survived on average 45 days (range, 29-64; P < 0.05). CONCLUSIONS Our data indicate that recipient-matching of passenger leukocytes significantly prolongs lung allograft survival.
Collapse
|
12
|
Madariaga MLL, Spencer PJ, Michel SG, La Muraglia GM, O’Neil MJ, Mannon EC, Leblang C, Rosales IA, Colvin RB, Sachs DH, Allan JS, Madsen JC. Effects of Lung Cotransplantation on Cardiac Allograft Tolerance Across a Full Major Histocompatibility Complex Barrier in Miniature Swine. Am J Transplant 2016; 16:979-86. [PMID: 26469344 PMCID: PMC5010442 DOI: 10.1111/ajt.13489] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/10/2015] [Accepted: 08/09/2015] [Indexed: 01/25/2023]
Abstract
A 12-day course of high-dose tacrolimus induces tolerance of major histocompatibility complex-mismatched lung allografts in miniature swine but does not induce tolerance of heart allografts unless a kidney is cotransplanted. To determine whether lungs share with kidneys the ability to induce cardiac allograft tolerance, we investigated heart-lung cotransplantation using the same induction protocol. Hearts (n = 3), heart-kidneys (n = 3), lungs (n = 6), and hearts-lungs (n = 3) were transplanted into fully major histocompatibility complex-mismatched recipients treated with high-dose tacrolimus for 12 days. Serial biopsy samples were used to evaluate rejection, and in vitro assays were used to detect donor responsiveness. All heart-kidney recipients and five of six lung recipients demonstrated long-term graft survival for longer than 272 days, while all heart recipients rejected their allografts within 35 days. Tolerant recipients remained free of alloantibody and showed persistent donor-specific unresponsiveness by cell-mediated lympholysis/mixed-lymphocyte reaction. In contrast, heart-lung recipients demonstrated rejection of both allografts (days 47, 55, and 202) and antidonor responsiveness in vitro. In contrast to kidneys, lung cotransplantation leads to rejection of both heart and lung allografts, indicating that lungs do not have the same tolerogenic capacity as kidneys. We conclude that cells or cell products present in kidney, but not heart or lung allografts, have a unique capacity to confer unresponsiveness on cotransplanted organs, most likely by amplifying host regulatory mechanisms.
Collapse
Affiliation(s)
- M. L. L. Madariaga
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - P. J. Spencer
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - S. G. Michel
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - G. M. La Muraglia
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - M. J. O’Neil
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - E. C. Mannon
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - C. Leblang
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - I. A. Rosales
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - R. B. Colvin
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - D. H. Sachs
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | | | | |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW When it comes to tolerance induction, kidney allografts behave differently from heart allografts that behave differently from lung allografts. Here, we examine how and why different organ allografts respond differently to the same tolerance induction protocol. RECENT FINDINGS Allograft tolerance has been achieved in experimental and clinical kidney transplantation. Inducing tolerance in experimental recipients of heart and lung allografts has, however, proven to be more challenging. New protocols being developed in nonhuman primates based on mixed chimerism and cotransplantation of tolerogenic organs may provide mechanistic insights to help overcome these challenges. SUMMARY Tolerance induction protocols that are successful in patients transplanted with 'tolerance-prone' organs such as kidneys and livers will most likely not succeed in recipients of 'tolerance-resistant' organs such as hearts and lungs. Separate clinical trials using more robust tolerance protocols will be required to achieve tolerance in heart and lung recipients.
Collapse
|
14
|
De Vleeschauwer S, Vanaudenaerde B, Vos R, Meers C, Wauters S, Dupont L, Van Raemdonck D, Verleden G. The need for a new animal model for chronic rejection after lung transplantation. Transplant Proc 2014; 43:3476-85. [PMID: 22099823 DOI: 10.1016/j.transproceed.2011.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The single most important cause of late mortality after lung transplantation is obliterative bronchiolitis (OB), clinically characterized by a decrease in lung function and morphologically by characteristic changes. Recently, new insights into its pathogenesis have been acquired: risk factors have been identified and the use of azithromycin showed a dichotomy with at least 2 different phenotypes of bronchiolitis obliterans syndrome (BOS). It is clear that a good animal model is indispensable to further dissect and unravel the pathogenesis of BOS. Many animal models have been developed to study BOS but, so far, none of these models truly mimics the human situation. Looking at the definition of BOS, a good animal model implies histological OB lesions, possibility to measure lung function, and airway inflammation. This review sought to discuss, including pros and cons, all potential animal models that have been developed to study OB/BOS. It has become clear that a new animal model is needed; recent developments using an orthotopic mouse lung transplantation model may offer the answer because it mimics the human situation. The genetic variants among this species may open new perspectives for research into the pathogenesis of OB/BOS.
Collapse
Affiliation(s)
- S De Vleeschauwer
- Laboratory of Pneumology, Kathoholieke Universiteit Leuven and UZ Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Tonsho M, Michel S, Ahmed Z, Alessandrini A, Madsen JC. Heart transplantation: challenges facing the field. Cold Spring Harb Perspect Med 2014; 4:4/5/a015636. [PMID: 24789875 DOI: 10.1101/cshperspect.a015636] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There has been significant progress in the field of heart transplantation over the last 45 years. The 1-yr survival rates following heart transplantation have improved from 30% in the 1970s to almost 90% in the 2000s. However, there has been little change in long-term outcomes. This is mainly due to chronic rejection, malignancy, and the detrimental side effects of chronic immunosuppression. In addition, over the last decade, new challenges have arisen such as increasingly complicated recipients and antibody-mediated rejection. Most, if not all, of these obstacles to long-term survival could be prevented or ameliorated by the induction of transplant tolerance wherein the recipient's immune system is persuaded not to mount a damaging immune response against donor antigens, thus eliminating the need for chronic immunosuppression. However, the heart, as opposed to other allografts like kidneys, appears to be a tolerance-resistant organ. Understanding why organs like kidneys and livers are prone to tolerance induction, whereas others like hearts and lungs are tolerance-resistant, could aid in our attempts to achieve long-term, immunosuppression-free survival in human heart transplant recipients. It could also advance the field of pig-to-human xenotransplantation, which, if successful, would eliminate the organ shortage problem. Of course, there are alternative futures to the field of heart transplantation that may include the application of total mechanical support, stem cells, or bioengineered whole organs. Which modality will be the first to reach the ultimate goal of achieving unlimited, long-term, circulatory support with minimal risk to longevity or lifestyle is unknown, but significant progress in being made in each of these areas.
Collapse
Affiliation(s)
- Makoto Tonsho
- MGH Transplantation Center, Massachusetts General Hospital, Boston, Massachusetts 02114
| | | | | | | | | |
Collapse
|
16
|
Jungraithmayr W, Jang JH, Schrepfer S, Inci I, Weder W. Small Animal Models of Experimental Obliterative Bronchiolitis. Am J Respir Cell Mol Biol 2013; 48:675-84. [DOI: 10.1165/rcmb.2012-0379tr] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
17
|
Sui H, Olivier AK, Klesney-Tait JA, Brooks L, Tyler SR, Sun X, Skopec A, Kline J, Sanchez PG, Meyerholz DK, Zavazava N, Iannettoni M, Engelhardt JF, Parekh KR. Ferret lung transplant: an orthotopic model of obliterative bronchiolitis. Am J Transplant 2013; 13:467-73. [PMID: 23205765 PMCID: PMC3638989 DOI: 10.1111/j.1600-6143.2012.04337.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/25/2012] [Accepted: 10/14/2012] [Indexed: 01/25/2023]
Abstract
Obliterative bronchiolitis (OB) is the primary cause of late morbidity and mortality following lung transplantation. Current animal models do not reliably develop OB pathology. Given the similarities between ferret and human lung biology, we hypothesized an orthotopic ferret lung allograft would develop OB. Orthotopic left lower lobe transplants were successfully performed in 22 outbred domestic ferrets in the absence of immunosuppression (IS; n = 5) and presence of varying IS protocols (n = 17). CT scans were performed to evaluate the allografts. At intervals between 3-6 months the allografts were examined histologically for evidence of acute/chronic rejection. IS protects allografts from acute rejection and early graft loss. Reduction of IS dosage by 50% allowed development of controlled rejection. Allografts developed infiltrates on CT and classic histologic acute rejection and lymphocytic bronchiolitis. Cycling of IS, to induce repeated episodes of controlled rejection, promoted classic histologic hallmarks of OB including fibrosis-associated occlusion of the bronchiolar airways in all allografts of long-term survivors. In conclusion, we have developed an orthotopic lung transplant model in the ferret with documented long-term functional allograft survival. Allografts develop acute rejection and lymphocytic bronchiolitis, similar to humans. Long-term survivors develop histologic changes in the allografts that are hallmarks of OB.
Collapse
Affiliation(s)
- H. Sui
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - A. K. Olivier
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - J. A. Klesney-Tait
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - L. Brooks
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - S. R. Tyler
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - X. Sun
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - A. Skopec
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - J. Kline
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - P. G. Sanchez
- Department of Cardiothoracic Surgery, University of Maryland, Baltimore, MD
| | - D. K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - N. Zavazava
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - M. Iannettoni
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - J. F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - K. R. Parekh
- Department of Cardiothoracic Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
18
|
Meltzer AJ, Veillette GR, Aoyama A, Kim KM, Cochrane ME, Wain JC, Madsen JC, Sachs DH, Rosengard BR, Allan JS. Donor brain death inhibits tolerance induction in miniature swine recipients of fully MHC-disparate pulmonary allografts. Am J Transplant 2012; 12:1290-5. [PMID: 22300400 PMCID: PMC3648993 DOI: 10.1111/j.1600-6143.2011.03949.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have previously shown that a short course of high-dose tacrolimus induces long-term tolerance to fully mismatched lung allografts procured from healthy MHC-inbred miniature swine. Here, we investigate whether donor brain death affects tolerance induction. Four recipient swine were transplanted with fully mismatched lung grafts from donors that were rendered brain dead and mechanically ventilated for 4 h before procurement (Group 1). These recipients were compared to two control groups (Group 2: 4 h of donor ventilation without brain death [n = 5]; and Group 3: no donor brain death with <1 h of ventilation [n = 6]). All recipients were treated with a 12-day course of tacrolimus. In contrast to both groups of control animals, the swine transplanted with lung allografts from brain dead donors all rejected their grafts by postoperative day 45 and showed persistent responsiveness to donor antigen by MLR. Several additional swine underwent brain death induction and/or mechanical ventilation alone to determine the effects of these procedures on the expression of proinflammatory molecules. Significant increases in serum concentrations of IL-1, TNF-α and IL-10 were seen after brain death. Upregulation of IL-1 and IL-6 gene expression was also observed.
Collapse
|
19
|
Beneficial effects of perioperative low-dose inhaled carbon monoxide on pulmonary allograft survival in MHC-inbred CLAWN miniature swine. Transplantation 2011; 90:1336-43. [PMID: 21076382 DOI: 10.1097/tp.0b013e3181ff8730] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND We have recently reported that perioperative low-dose carbon monoxide (CO) inhalation decreases lung ischemia-reperfusion injury in miniature swine. The aims of this study were to establish a large animal model of pulmonary allograft rejection using polymerase chain reaction-typed major histocompatibility complex (MHC)-inbred CLAWN miniature swine and to examine the effects of CO on allograft survival. METHODS Eleven CLAWN miniature swines received fully MHC-mismatched lungs followed by 12 days of tacrolimus (days 0-11; blood level 35-45 ng/mL). Six recipients received tacrolimus alone (control group). Five recipients were additionally treated with inhaled CO (180 min for donors until graft harvest; 390 min for recipients until 2 hr after reperfusion). RESULTS All recipients treated with tacrolimus alone uniformly rejected their grafts by postoperative day 63 with development of cytotoxic antidonor antibodies. CO treatment was effective in prolonging allograft survival from a mean of 47±7 to 82±13 days (P=0.017), with one CO-treated animal maintaining function until postoperative day 120. Development of antidonor antibodies and donor-specific responsiveness by cell-mediated lympholysis and mixed lymphocyte reaction assays was delayed in animals that received CO therapy. Furthermore, serum concentrations of proinflammatory cytokines (interleukin-1β and -6) 1 day after transplant were significantly decreased in the CO-treated group. CONCLUSIONS Fully MHC-mismatched lungs in CLAWN miniature swine were consistently rejected within 63 days, suggesting that this is a robust large animal model ideal for investigating mechanisms and treatment of lung rejection. Perioperative low-dose CO inhalation prolonged graft survival and inhibited antidonor antibody production and was associated with decreased proinflammatory mediators in this model.
Collapse
|
20
|
Animal models of chronic allograft injury: contributions and limitations to understanding the mechanism of long-term graft dysfunction. Transplantation 2010; 90:935-44. [PMID: 20703180 DOI: 10.1097/tp.0b013e3181efcfbc] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Advances in immunosuppression have reduced the incidence of acute graft loss after transplantation, but long-term allograft survival is still hindered by the development of chronic allograft injury, a multifactorial process that involves both immunologic and nonimmunologic components. Because these components become defined in the clinical setting, development of animal models enables exploration into underlying mechanisms leading to long-term graft dysfunction. This review presents animal models that have enabled investigation into chronic allograft injury and discusses pivotal models currently being used. The mechanisms uncovered by these models will ultimately lead to development of new therapeutic options to prevent long-term graft dysfunction.
Collapse
|
21
|
|
22
|
Sato M, Keshavjee S, Liu M. Translational research: animal models of obliterative bronchiolitis after lung transplantation. Am J Transplant 2009; 9:1981-7. [PMID: 19663891 DOI: 10.1111/j.1600-6143.2009.02770.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Obliterative bronchiolitis (OB) or chronic graft dysfunction remains the major limitation to long-term success of lung transplantation. Investigation using animal models is a critical component of research to understand the underlying pathological mechanisms and to develop novel preventive and therapeutic strategies for OB. Multiple animal models of OB exist, including orthotopic lung transplantation in rodents and large animals, orthotopic tracheal transplantation and heterotopic transplantation of a trachea in variable sites such as subcutaneous, intraomental and intrapulmonary sites. The most important issue for researchers is not specifically which model is the best but which is the most appropriate model to test their scientific hypothesis. For example, while orthotopic lung transplantation best mimics the overall surgical procedure, a question regarding fibrotic processes of OB may be better answered using heterotopic tracheal transplant models because of their reliable reproducibility of allograft obliterative airway fibrosis. Animal models should be continuously refined, modified and sometimes combined to fit the particular research purpose. We review the available animal models, their modifications and possible applications to assist researchers in choosing the appropriate model for their intended research.
Collapse
Affiliation(s)
- M Sato
- Latner Thoracic Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
23
|
Satoda N, Shoji T, Wu Y, Fujinaga T, Chen F, Aoyama A, Zhang JT, Takahashi A, Okamoto T, Matsumoto I, Sakai H, Li Y, Zhao X, Manabe T, Kobayashi E, Sakaguchi S, Wada H, Ohe H, Uemoto S, Tottori J, Bando T, Date H, Koshiba T. Value of FOXP3 expression in peripheral blood as rejection marker after miniature swine lung transplantation. J Heart Lung Transplant 2009; 27:1293-301. [PMID: 19059109 DOI: 10.1016/j.healun.2008.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Revised: 08/01/2008] [Accepted: 08/21/2008] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Outcome for highly immunogenic lung transplantation remains unsatisfactory despite the development of potent immunosuppressants. The poor outcome may be the result of a lack of minimally invasive methods to detect early rejection. There is emerging clinical evidence that, paradoxically, expression of forkhead box P3 (FOXP3, a specific marker for the regulatory T cells) is upregulated within rejecting grafts. METHODS Orthotopic lung transplantation was performed using miniature swine without immunosuppression. Rejection was monitored by chest radiography and open lung biopsy. Expressions levels of FOXP3, perforin, Fas-L and IP-10 mRNA were quantified in the peripheral blood. In addition, rescue immunosuppressive therapy (steroid plus tacrolimus) was administered on post-operative day (POD) 4 or 6. RESULTS Early rejection was detected by open lung biopsy, but misdiagnosed by chest radiography on POD 4. Expression of FOXP3 in the peripheral blood reached its highest value as early as POD 4, followed by a decline. Such an increase of FOXP3 was not observed in recipients given high-dose tacrolimus. Neither perforin, Fas-L or IP-10 in the peripheral blood exhibited significant fluctuations in the early phase of rejection. Rescue immunosuppressive therapy from POD 4, when peak FOXP3 was seen, prolonged graft survival (27.2 days, versus 9.1 days without immunosuppression, p < 0.001), in contrast to POD 6, when rejection was suspected by chest radiography (11.5 days, p = not statistically significant [NS]). CONCLUSIONS In a miniature swine lung transplantation model, the FOXP3 mRNA level in the peripheral blood was upregulated at an early phase of rejection. The clinical implication of this finding remains to be elucidated.
Collapse
Affiliation(s)
- Naoki Satoda
- Department of Thoracic Surgery, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Weiss MJ, Guenther DA, Mezrich JD, Sahara H, Ng CY, Meltzer AJ, Sayre JK, Cochrane ME, Pujara AC, Houser SL, Sachs DH, Rosengard BR, Allan JS, Benichou G, Madsen JC. The indirect alloresponse impairs the induction but not maintenance of tolerance to MHC class I-disparate allografts. Am J Transplant 2009; 9:105-13. [PMID: 19145702 PMCID: PMC3252388 DOI: 10.1111/j.1600-6143.2008.02494.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We studied the effects of indirect allorecognition on the induction and maintenance phases of tolerance in miniature swine cotransplanted with heart and kidney allografts. MHC class I-mismatched heart and kidney grafts were cotransplanted in recipients receiving CyA for 12 days. Recipients were unimmunized or immunized with a set of donor-derived or control third-party MHC class I peptides either 21 days prior to transplantation or over 100 days after transplantation. T-cell proliferation, delayed type hypersensitivity reaction (DTH) and antibody production were assessed. All animals injected with donor MHC class I peptides developed potent indirect alloresponses specific to the immunizing peptides. While untreated recipients developed stable tolerance, all animals preimmunized with donor allopeptides rejected kidney-heart transplants acutely. In contrast, when peptide immunization was delayed until over 100 days after kidney-heart transplantation, no effects were observed on graft function or in vitro measures of alloimmunity. Donor peptide immunization prevented tolerance when administered to recipients pre transplantation but did not abrogate tolerance when administered to long-term survivors post transplantation. This suggests that the presence of T cells activated via indirect allorecognition represent a barrier to the induction but not the maintenance of tolerance.
Collapse
Affiliation(s)
- Matthew J. Weiss
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dax A. Guenther
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Joshua D. Mezrich
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hisashi Sahara
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Choo Y. Ng
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Andrew J. Meltzer
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jessica K. Sayre
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Meghan E. Cochrane
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Akshat C. Pujara
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Stuart L. Houser
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - David H. Sachs
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Bruce R. Rosengard
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - James S. Allan
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston,Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Gilles Benichou
- Division of Abdominal Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Joren C. Madsen
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston,Address correspondence to: Joren C. Madsen, M.D., D.Phil., Cox 654, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114.
| |
Collapse
|
25
|
Operational tolerance to class I disparate lungs can be induced despite pretransplant immunization with class I allopeptides. Transplantation 2007; 84:1467-73. [PMID: 18091523 DOI: 10.1097/01.tp.0000288321.67926.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Using a class I-disparate swine lung transplant model, we examined whether an intensive course of tacrolimus could induce operational tolerance and whether preoperative allopeptide immunization would prevent the development of tolerance. METHODS Left lung grafts were performed using class I-disparate (class II-matched) donors. Recipients were treated with 12 days of postoperative tacrolimus. Three recipients were immunized prior to transplantation with class I allopeptides. Three other recipients were not immunized. RESULTS The nonimmunized recipients maintained their grafts long term (>497, >451, and >432 days), without developing chronic rejection. The immunized swine also maintained their grafts long term (>417, >402, >401 days), despite developing a variety of in vitro and in vivo responses to the immunizing peptides, as well as having strong mixed lymphocyte reactions to donor cells prior to transplantation. CONCLUSIONS Using only a brief course of tacrolimus, we have been able to induce a state of operational tolerance in a class I-disparate preclinical lung transplant model. Moreover, preoperative alloimmunization did not block tolerance induction or induce chronic rejection. These data show that it is possible to create a state of operational tolerance to lung allografts even in the presence of donor-sensitized cells.
Collapse
|
26
|
Wang WL, Yao MY, Jin J, Jia CK, Gao LH, Xie HY, Zheng SS. Increased expression of non-interleukin-2 T cell growth factors and their implications during liver allograft rejection in rats. J Gastroenterol Hepatol 2007; 22:1141-7. [PMID: 17532783 DOI: 10.1111/j.1440-1746.2007.04925.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Rejection remains a problem in the transplantation field. The aim of this study was to establish acute and chronic rejection models in rats and to investigate the roles of non-interleukin (IL)-2 T cell growth factors such as IL-15, IL-7 and IL-13 during rejection. METHODS A liver transplant model was established using Dark Agouti and Brown Norway rats. The rats were divided into group A, left without treatment; group B, received cyclosporinee (1 mg/kg/day); and group C, cyclosporinee (4 mg/kg/day). Histopathological, reverse transcriptase-polymerase chain reaction and western blot were performed in liver specimens obtained from different time-points after transplantation in the three groups. RESULTS In group A, the livers showed irreversible acute cellular rejection with cell infiltration. In group B, chronic liver rejection was found, with graft infiltration, ductular damage or proliferation, obliterative arteriopathy and liver fibrosis. No apparent histological alterations were observed in group C. IL-15, IL-7 and IL-13 messenger RNA and their protein were all highly expressed in the liver specimens of groups A and B. Upregulated expression was found in IL-15 since the first day after transplantation and in IL-7 and IL-13 since day 6. The extent of IL-15 upregulation was more than that of IL-7 and IL-13. CONCLUSIONS Liver transplantation in Dark Agouti to Brown Norway rats with low-dose immunosuppression can induce chronic rejection. In the process of acute and chronic allograft rejections, non-IL-2 T cell growth factors such as IL-15, IL-7 and IL-13 play roles. Strategies should pay more attention to regulating these cytokines after liver transplantation.
Collapse
Affiliation(s)
- Wei-Lin Wang
- Department of Hepatobiliary Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital of Medical College, Zhejiang University, Hang Zhou, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Wakeman DR, Crain AM, Snyder EY. Large animal models are critical for rationally advancing regenerative therapies. Regen Med 2007; 1:405-13. [PMID: 17465832 PMCID: PMC2905042 DOI: 10.2217/17460751.1.4.405] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Dustin R Wakeman
- Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
| | - Andrew M Crain
- Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
| | - Evan Y Snyder
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
- Correspondence: ; 858-646-3158 Fax: 858-713-6273
| |
Collapse
|
28
|
Sahara H, Weiss MJ, Ng CY, Houser SL, Pujara AC, Sayre JK, Wain JC, Sachs DH, Madsen JC, Allan JS. Thymectomy does not abrogate long-term acceptance of MHC class I-disparate lung allografts in miniature Swine. Transplant Proc 2007; 38:3253-5. [PMID: 17175240 PMCID: PMC1865569 DOI: 10.1016/j.transproceed.2006.10.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Indexed: 10/23/2022]
Abstract
UNLABELLED We have previously reported that tolerance to class I disparate lung allografts in miniature swine could be induced using an intensive 12-day course of tacrolimus and that pretransplant sensitization with immunogenic MHC class I allopeptides failed to block the induction of tolerance. We also have previously reported the importance of the presence of the thymus in the induction of tolerance to isolated heart, kidney, and combined heart-kidney transplants. In this study, we examined the impact of thymectomy on tolerance induction in lung transplantation. METHODS Orthotopic left lung transplantation was performed using MHC class I-disparate donors. The recipients received a 12-day course of high-dose tacrolimus (n = 6). Total thymectomies were performed in three of the swine 21 days prior to transplantation. Lung grafts were monitored by chest radiography and serial open lung biopsy. RESULTS All euthymic recipients maintained their grafts for over 1 year. None of the thymectomized recipients has experienced graft loss in the 6 to 10 months following transplantation. Although isolated lesions of obliterative bronchiolitis were occasionally seen in one thymectomized animal on biopsy, donor-specific unresponsiveness has been observed on assays of cell-mediated lymphocytotoxicity in all recipients. Moreover, co-culture assays have shown that recipient lymphocytes can strongly inhibit the normally robust response of naïve recipient-matched lymphocytes to donor antigen. This inhibition was not seen when using stimulators primed with third-party antigens against appropriate targets. CONCLUSIONS These data suggest that thymus-independent peripheral regulatory mechanisms may be sufficient to induce and maintain long-term acceptance of the lung allografts.
Collapse
Affiliation(s)
- H Sahara
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Shoji T, Sahara H, Muniappan A, Guenther DA, Houser SL, Pujara AC, Bravard MA, Wain JC, Sachs DH, Madsen JC, Allan JS. An MHC class II disparity raises the threshold for tolerance induction in pulmonary allografts in miniature swine. Transplant Proc 2007; 38:3268-70. [PMID: 17175245 PMCID: PMC1766937 DOI: 10.1016/j.transproceed.2006.10.161] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The mechanisms and treatment of chronic rejection in pulmonary allotransplantation remain elusive. We have induced robust tolerance to class I-disparate lung allografts in miniature swine using an intensive 12-day course of tacrolimus. Here, we tested whether a tolerant state can be induced in swine receiving fully mismatched lung allografts. METHODS Orthotopic left lung allografts were performed using MHC class I-disparate (group 1: n = 3) or fully disparate (group 2: n = 6) donors. The recipients received a 12-day postoperative course of tacrolimus (continuous intravenous infusion; target level = 35-50 ng/mL) as their only immunosuppression. RESULTS All swine in group 1 maintained their grafts long term without developing any lesions of chronic rejection (>497, >432, >451 days). These recipients exhibited donor-specific hyporesponsiveness in cell-mediated lymphocytotoxity (CML) and mixed lymphocyte reaction (MLR) assays. In group 2, five of the six recipients maintained their grafts long term (sacrificed on postoperative days 515, 389, 429, 481, and 438 with viable grafts). Isolated lesions of obliterative bronchiolitis were occasionally seen on biopsy, and donor-specific hyporesponsiveness on assays was consistently observed. The remaining recipient rejected its graft on day 103 with histologic findings of obliterative bronchiolitis. CONCLUSIONS We report long-term graft acceptance without chronic immunosuppression in five of six recipients across a full MHC disparity, albeit with some evidence of obliterative bronchiolitis. These data suggest that the class II disparity inherent in a fully mismatched transplant increases the requirement for tolerance induction.
Collapse
Affiliation(s)
- Tsuyoshi Shoji
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Hisashi Sahara
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ashok Muniappan
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Dax A. Guenther
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Stuart L. Houser
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Akshat C. Pujara
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Marjory A. Bravard
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - John C. Wain
- Division of Thoracic Surgery; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - David H. Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Joren C. Madsen
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Division of Cardiac Surgery; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - James S. Allan
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Division of Thoracic Surgery; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Address correspondence to: James S. Allan, MD, Massachusetts General Hospital, Blake 1570, 55 Fruit Street, Boston, MA 02114. Fax: 978-745-9024, Phone: 617-724-1103, E-mail:
| |
Collapse
|
30
|
Sahara H, Shoji T, Ng CY, Weiss MJ, Muniappan A, Guenther DA, Houser SL, Pujara AC, Sayre JK, Wain JC, Sachs DH, Madsen JC, Allan JS. The role of indirect recognition of MHC class I and II allopeptides in a fully mismatched miniature swine model of lung transplantation. Transplant Proc 2007; 38:3256-8. [PMID: 17175241 PMCID: PMC1865568 DOI: 10.1016/j.transproceed.2006.10.059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Indexed: 11/16/2022]
Abstract
UNLABELLED Considerable evidence suggests that indirect recognition of MHC allopeptides plays an important role in solid-organ rejection. Here, we examine whether immunization with class I or class II allopeptides accelerates rejection in a fully MHC-mismatched lung transplant model in miniature swine. METHODS Recipients were immunized with either donor-derived class I or class II peptides. Sensitization to the peptides was confirmed by DTH testing and in vitro proliferation assays. Nonimmunized control (n = 6), class I peptide-immunized (n = 3), and class II peptide-immunized (n = 3) swine were transplanted with fully mismatched lungs using only a 12-day course of tacrolimus. RESULTS One control animal rejected its graft on postoperative day 103, while the others maintained their grafts for over 1 year. In the class I peptide-immunized group, two recipients rejected their grafts (days 14 and 52). The third animal has not rejected the graft (day 120, experiment is ongoing). In contrast, in the class II-peptide immunized group, only one animal rejected its graft on day 52, while the others maintained their grafts over 1 year. Both anti-donor IgM and IgG antibodies were detectable in all acute rejectors, although no alloantibody was detectable in long-term acceptors. Regardless of the fate of the graft, all animals have maintained their proliferative responses to the peptides. However, only acceptors maintained donor-specific hyporesponsiveness in cell-mediated lymphocytotoxity and mixed lymphocyte reaction assays. CONCLUSIONS Pretransplant sensitization of lung allograft recipients to donor allopeptides accelerates graft rejection. This appears particularly true for class I-derived allopeptides, suggesting that class II molecules may be less antigenic when presented indirectly.
Collapse
Affiliation(s)
- H Sahara
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Obliterative bronchiolitis is a devastating illness that limits the long-term success of lung transplantation. Its high prevalence and overall poor response to current therapeutic measures demands further research to elucidate pathogenic mechanisms. Toward this goal, there is a role for animal models to study the mechanisms of obliterative bronchiolitis, such as the murine heterotopic tracheal allograft model. This review compares the tracheal allograft model to human obliterative bronchiolitis pathology and highlights the important mechanisms of airway rejection described using this model. Although certain limitations exist, the pursuit of proof-of-concept studies in this model, as well as other animal models, can provide the basis for genetic and cellular translational human studies directed toward post-transplant obliterative bronchiolitis pathogenesis. To meet these challenges, we call for the establishment of a National Institutes of Health-supported Lung Transplant Network to better orchestrate translational research efforts in obliterative bronchiolitis pathogenesis and treatment, and to advance the field of lung transplantation.
Collapse
Affiliation(s)
- John F McDyer
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
32
|
Warnecke G, Avsar M, Morancho M, Peters C, Thissen S, Kruse B, Baumann R, Ungefroren H, Simon AR, Hohlfeld JM, Karstens JH, Haverich A, Strüber M. Preoperative Low-Dose Irradiation Promotes Long-Term Allograft Acceptance and Induces Regulatory T Cells in a Porcine Model of Pulmonary Transplantation. Transplantation 2006; 82:93-101. [PMID: 16861947 DOI: 10.1097/01.tp.0000225833.23093.ed] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND A simplified conditioning protocol including single-dose preoperative thymic and low-dose whole body irradiation with or without subsequent donor bone marrow transplantation (BMTx) can be applied in porcine lung transplantation. We hypothesized that this protocol would prolong allograft survival. METHODS Left-sided single lung transplantation from major histocompatibility complex (MHC)-mismatched donors was performed in 27 minipigs. Recipients received whole body (1.5 Gy) and thymic irradiation (7 Gy) before transplantation (IRR group, n=6), intravenous immunosuppression with methylprednisolone, cyclosporine, and azathioprine for 27 postoperative days (IS group, n=5) or both (IRR+IS group, n=10). BMTx group recipients were treated with irradiation, immunosuppression and a donor bone marrow infusion on postoperative day 1. Peripheral blood leukocyte phenotype and donor cell chimerism were monitored by flow cytometry. Purified CD25+ T cells from long-term survivors or rejecting animals were used for in vitro MLR suppression assays. RESULTS Median graft survival was: IRR 12 days, IS 55 days, IRR+IS 239 days, and BMTx 80 days (P<0.0001). Early peripheral blood macrochimerism was substantial in both the IRR+IS and the BMTX group but was lost in all groups after day 80. The frequency and suppressive function of CD4+CD25+ T cells were enhanced in IRR+IS group long-term survivors. CONCLUSION Although donor bone marrow infusion was not beneficial in our model, a substantial proportion of the animals treated with irradiation and a 28-day course of immunosuppression accepted their lung allografts long term. The mechanism involved in maintaining allograft tolerance may be based on peripheral T-cell regulation.
Collapse
Affiliation(s)
- Gregor Warnecke
- Division of Thoracic and Cardiovascular Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Neuringer IP, Chalermskulrat W, Aris R. Obliterative bronchiolitis or chronic lung allograft rejection: a basic science review. J Heart Lung Transplant 2005; 24:3-19. [PMID: 15653373 DOI: 10.1016/j.healun.2004.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2003] [Revised: 12/12/2003] [Accepted: 01/06/2004] [Indexed: 01/06/2023] Open
Affiliation(s)
- Isabel P Neuringer
- Division of Pulmonary and Critical Care Medicine and Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.
| | | | | |
Collapse
|
34
|
Gao LH, Zheng SS, Zhu YF, Wan YL, Wang YL, Wei GQ, Qian SK, Qian SQ, Jiang WJ. A rat model of chronic allograft liver rejection. Transplant Proc 2005; 37:2327-32. [PMID: 15964409 DOI: 10.1016/j.transproceed.2005.03.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2005] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The aim of this study was to develop a rat model of chronic irreversible rejection, which is a major causes of late graft loss and retransplantation after orthotopic liver allotransplantation. METHODS Allogeneic liver transplantation was performed in a rat combination of Dark Agouti (DA) to Brown Norway (BN). Group A was left without treatment, group B received cyclosporine' (CsA; 1 mg/kg/d) and group C, CsA (4 mg/kg/d). Animals were followed for 6 months. Liver tissue was harvested to construct a time course of histological changes after liver transplantation using histopathological and morphometric techniques. We compared the total histological score of rejection activity index and survival rates. RESULTS In untreated animals, irreversible acute rejection developed, all animals died within 15 days. In the low-dose CsA group, all animals that survived more than 30 days developed moderate to severe manifestations of chronic liver rejection, with graft infiltration, ductular damage or proliferation, obliterative arteriopathy, and liver fibrosis. No apparent histological alterations were observed in group C. Survival analysis showed significant differences between the three groups. CONCLUSIONS In the rat strain combination of DA --> BN with low-dose immunosuppression, early mild inflammation was followed by the development of chronic rejection.
Collapse
Affiliation(s)
- L H Gao
- Dept of Hepatobiliary and Pancreatic Surgery, Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road, 79 Hangzhou 310003, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shoji T, Wain JC, Houser SL, Benjamin LC, Johnston DR, Hoerbelt R, Hasse RS, Lee RS, Muniappan A, Guenther DA, Bravard MA, Ledgerwood LG, Sachs DH, Sayegh MH, Madsen JC, Allan JS. Indirect recognition of MHC class I allopeptides accelerates lung allograft rejection in miniature swine. Am J Transplant 2005; 5:1626-34. [PMID: 15943620 DOI: 10.1111/j.1600-6143.2005.00925.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The role of indirect allorecognition in graft rejection is examined in two experiments using a swine lung transplantation model. First, two swine received class I mismatched grafts without immunosuppression; another two recipients were treated postoperatively with cyclosporine (CsA). These swine exhibited acute and chronic rejection, respectively. All four recipients developed T-cell reactivity to donor-derived class I major histocompatibility complex (MHC) peptides. Second, six swine were immunized with synthetic donor-derived class I allopeptides prior to transplantation. Control groups consisted of nonimmunized recipients (n = 6) and recipients immunized with an irrelevant peptide (n = 3). These recipients all received a 12-day course of post-operative CsA. Swine immunized with allopeptides exhibited accelerated graft rejection, as compared to both control groups (p < 0.01 and p = 0.03, respectively). Within the experimental group, the dominant histologic finding was acute rejection (AR). Obliterative bronchiolitis (OB) was seen in the graft with the longest survival. Both control groups showed a lesser degree of AR, with four out of six nonimmunized swine ultimately developing OB. These studies suggest that indirect allorecognition is operative during lung allograft rejection, and that pre-transplant sensitization to donor-derived MHC allopeptides can accelerate graft rejection.
Collapse
Affiliation(s)
- Tsuyoshi Shoji
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Madsen JC. Is thoracic organ transplantation ready for tolerance? Transplant Rev (Orlando) 2005. [DOI: 10.1016/j.trre.2005.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Shoji T, Muniappan A, Guenther DA, Wain JC, Houser SL, Hoerbelt R, Benjamin LC, Hasse RS, Bravard MA, Sachs DH, Madsen JC, Allan JS. Long-term acceptance of porcine pulmonary allografts without chronic rejection. Transplant Proc 2005; 37:72-4. [PMID: 15808551 DOI: 10.1016/j.transproceed.2004.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVES The mechanisms and treatment of chronic rejection in pulmonary allotransplantation remain elusive. Using a strategy to induce tolerance across strong allogeneic barriers, we have employed a brief, intensive course of immunosuppression to determine whether the induction of donor-specific hyporesponsiveness would prevent allograft rejection in a preclinical model of lung transplantation using MHC-inbred miniature swine. METHODS Orthotopic left lung allografts were performed using MHC class I-disparate donors. The recipients received a 12-day postoperative course of cyclosporine (n = 6) or a 12-day postoperative course of high-dose tacrolimus (n = 3) as their only immunosuppression. Control animals received no immunosuppression (n = 3). RESULTS Cyclosporine-treated recipients exhibited graft survival ranging from 67 to >605 days. All six animals developed acute cellular rejection between postoperative days (PODs) 27 and 108. Two animals lost their grafts on PODs 67 and 69, before developing obliterative bronchiolitis (OB). The other four recipients developed OB between PODs 119 and 238. In contrast, all tacrolimus-treated recipients maintained their grafts long term, without developing chronic rejection (>339, >308, and >231). These recipients also exhibited donor-specific hyporesponsiveness in assays of cell-mediated lymphocytotoxity. All untreated control animals lost their grafts to acute rejection by POD 11. CONCLUSIONS This study demonstrates the ability of a brief course of high-dose tacrolimus to induce long-term graft acceptance with donor-specific hyporesponsiveness in a class I-disparate preclinical lung transplant model.
Collapse
Affiliation(s)
- T Shoji
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Houser SL, Benjamin LC, Wain JC, Madsen JC, Allan JS. Constitutive expression of major histocompatibility complex class II antigens in pulmonary epithelium and endothelium varies among different species. Transplantation 2004; 77:605-7. [PMID: 15084944 DOI: 10.1097/01.tp.0000114285.63313.e7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We have observed high constitutive levels of class II antigen expression on porcine and human coronary endothelium, but not on the endothelium of rats and mice. This study examines whether a similar interspecies difference exists in the expression of class II molecules on pulmonary epithelium and endothelium. Lung tissues from naïve human, porcine, and rodent sources were stained with the monoclonal antibody ISCR3 and examined by light microscopy. Immunoperoxidase staining of class II molecules was observed on human and porcine pulmonary epithelium and endothelium, but was absent in rats and mice. By using an antibody with cross-species reactivity, we demonstrated that naïve swine pulmonary epithelium and endothelium, unlike those of rodent species, express basal levels of class II antigens in a manner similar to that observed in human lung tissue. These interspecies differences may explain experimental differences observed between murine and large-animal constructs.
Collapse
Affiliation(s)
- Stuart L Houser
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
39
|
|