1
|
Coppens A, Verleden SE, Claes E, Voet H, Verleden GM, Lapperre TS, Yildirim AÖ, Jungraithmayr W, Yamada Y, Peeters DJE, Hendriks JMH. Murine orthotopic lung transplant models: A comprehensive overview of genetic mismatch degrees and histopathological insights into chronic lung allograft dysfunction. Am J Transplant 2024; 24:1930-1940. [PMID: 39098448 DOI: 10.1016/j.ajt.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Currently, lung transplantation outcome remains inferior compared to other solid organ transplantations. A major cause for limited survival after lung transplantation is chronic lung allograft dysfunction. Numerous animal models have been developed to investigate chronic lung allograft dysfunction to discover adequate treatments. The murine orthotopic lung transplant model has been further optimized over the last years. However, different degrees of genetic mismatch between donor and recipient mice have been used, applying a single, minor, moderate, and major genetic mismatch. This review aims to reassess the existing murine mismatch models and provide a comprehensive overview, with a specific focus on their eventual histopathological presentation. This will be crucial to leverage this model and tailor it according to specific research needs.
Collapse
Affiliation(s)
- Axelle Coppens
- Department of Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, Wilrijk, Belgium; Department of Thoracic and Vascular Surgery, University Hospital of Antwerp, Edegem, Belgium
| | - Stijn E Verleden
- Department of Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, Wilrijk, Belgium; Department of Thoracic and Vascular Surgery, University Hospital of Antwerp, Edegem, Belgium; Department of Pulmonology, University Hospital of Antwerp, Edegem, Belgium
| | - Erik Claes
- Department of Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, Wilrijk, Belgium; Department of Thoracic and Vascular Surgery, University Hospital of Antwerp, Edegem, Belgium
| | - Hanne Voet
- Department of Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, Wilrijk, Belgium; Department of Pulmonology, University Hospital of Antwerp, Edegem, Belgium
| | - Geert M Verleden
- Department of Pulmonology, University Hospital of Antwerp, Edegem, Belgium
| | - Therese S Lapperre
- Department of Pulmonology, University Hospital of Antwerp, Edegem, Belgium; Laboratory of Experimental Medicine and Pediatrics, Division of Respiratory Medicine, University of Antwerp, Wilrijk, Belgium
| | - Ali Ö Yildirim
- Divison of Immunopathology in COPD, Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany; Division of Immunopathologu in COPD, Institute of Experimental Pneumology, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yoshito Yamada
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan; Department of Thoracic Surgery, Kyoto Katsura Hospital, Kyoto, Japan
| | - Dieter J E Peeters
- Department of Pathology, University Hospital of Antwerp, Edegem, Belgium
| | - Jeroen M H Hendriks
- Department of Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, Wilrijk, Belgium; Department of Thoracic and Vascular Surgery, University Hospital of Antwerp, Edegem, Belgium.
| |
Collapse
|
2
|
Mauer J, Kuckhahn A, Ramsperger-Gleixner M, Ensminger SM, Distler JH, Weyand M, Heim C. Nintedanib reduces alloimmune-induced chronic airway changes in murine tracheal allografts. Transpl Immunol 2022; 73:101608. [DOI: 10.1016/j.trim.2022.101608] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/12/2022] [Accepted: 04/23/2022] [Indexed: 01/01/2023]
|
3
|
Inhibiting S100A8/A9 attenuates airway obstruction in a mouse model of heterotopic tracheal transplantation. Biochem Biophys Res Commun 2022; 629:86-94. [DOI: 10.1016/j.bbrc.2022.08.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022]
|
4
|
Watanabe T, Juvet SC, Boonstra K, Guan Z, Joe B, Teskey G, Keshavjee S, Martinu T. Recipient bone marrow-derived IL-17 receptor A-positive cells drive allograft fibrosis in a mouse intrapulmonary tracheal transplantation model. Transpl Immunol 2021; 69:101467. [PMID: 34547417 DOI: 10.1016/j.trim.2021.101467] [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: 06/12/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
IL-17A is implicated in the pathogenesis of chronic lung allograft dysfunction, which limits survival after lung transplantation. While many cells express the IL-17 receptor A (IL-17RA) which is the main receptor for IL-17A, the cellular targets of IL-17A in development of post-transplant fibrosis are unknown. The purpose of this study was to determine whether IL-17RA expression by donor or recipient structural or bone marrow (BM) cells is required for the development of allograft fibrosis in a mouse intrapulmonary tracheal transplantation (IPTT) model. BM chimeras were generated using C57BL/6 and IL-17RA-knockout mice. After engraftment, allogeneic IPTTs were performed using the chimeric and BALB/c mice as donors or recipients. This allowed us to assess the effect of IL-17RA deficiency in recipient BM, recipient structural, donor BM, or donor structural compartments separately. Tracheal grafts, the surrounding lung, and mediastinal lymph nodes were assessed 28 days after IPTT. Only recipient BM IL-17RA deficiency resulted in attenuation of tracheal graft obliteration. In the setting of recipient BM IL-17RA deficiency, T cells and neutrophils were decreased in mediastinal lymph nodes. Additionally, recipient BM IL-17RA deficiency was associated with increased B220+PNAd+ lymphoid aggregates, consistent with tertiary lymphoid organs, in proximity to the tracheal allograft. In this IPTT model, recipient BM-derived cells appear to be the primary targets of IL-17RA signaling during fibrotic obliteration of the tracheal allograft.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, University Health Network, Canada; Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Stephen C Juvet
- Latner Thoracic Research Laboratories, University Health Network, Canada; Division of Respirology, Department of Medicine, University of Toronto, Canada
| | - Kristen Boonstra
- Latner Thoracic Research Laboratories, University Health Network, Canada
| | - Zehong Guan
- Latner Thoracic Research Laboratories, University Health Network, Canada
| | - Betty Joe
- Latner Thoracic Research Laboratories, University Health Network, Canada
| | - Grace Teskey
- Latner Thoracic Research Laboratories, University Health Network, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, University Health Network, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, University Health Network, Canada; Division of Respirology, Department of Medicine, University of Toronto, Canada.
| |
Collapse
|
5
|
Brugiere O, Verleden SE. Putting the spotlight on macrophage-derived cathepsin in the pathophysiology of obliterative bronchiolitis. Eur Respir J 2021; 57:57/5/2004607. [PMID: 33985982 DOI: 10.1183/13993003.04607-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/19/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Olivier Brugiere
- Lung Transplant Dept, Foch Hospital, Suresnes, France .,Inserm UMR S 1152, Physiopathologie et Epidémiologie des Maladies Respiratoires, Paris, France
| | | |
Collapse
|
6
|
Suzuki Y, Oishi H, Kanehira M, Matsuda Y, Hirama T, Noda M, Okada Y. Effect of CTLA4-Ig on Obliterative Bronchiolitis in a Mouse Intrapulmonary Tracheal Transplantation Model. Ann Thorac Cardiovasc Surg 2021; 27:355-365. [PMID: 33980752 PMCID: PMC8684841 DOI: 10.5761/atcs.oa.20-00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: One of the serious problems after lung transplantation is chronic lung allograft dysfunction (CLAD). Most CLAD patients pathologically characterized by obliterative bronchiolitis (OB). Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4)-Ig is a combination protein of the Fc fragment of human IgG1 linked to the extracellular domain of CTLA4. The aim of the study was to examine the effect of CTLA4-Ig therapy on OB using a mouse intrapulmonary tracheal transplantation (IPTT) model. Methods: IPTT was performed between BALB/c (donor) and C57BL/6 (recipient) mice. Abatacept, which is a commercially available form of CTLA4-Ig, was intraperitoneally injected in recipient mice immediately after surgery, on days 7, 14, and 21. The mice in the control group received human IgG. Results: We performed semi-quantitative analysis of graft luminal obliteration at post-transplant day 28. We calculated the obliteration ratio of the lumen of the transplanted trachea in each case. The obliteration ratio was significantly lower in the CTLA4-Ig group than that in the control group (91.2 ± 2.1% vs. 47.8 ± 7.9%, p = 0.0008). Immunofluorescent staining revealed significantly decreased lymphoid neogenesis in the lung. Conclusions: CTLA4-Ig therapy attenuated tracheal obliteration with fibrous tissue in the mouse IPTT model. The attenuation of fibrous obliteration was correlated with the inhibition of lymphoid neogenesis.
Collapse
Affiliation(s)
- Yamato Suzuki
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Hisashi Oishi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiko Kanehira
- Center for Life Science Research, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yasushi Matsuda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan.,Department of Thoracic Surgery, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Takashi Hirama
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Masafumi Noda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
7
|
Hennessy C, Lewik G, Cross A, Hester J, Issa F. Recent advances in our understanding of the allograft response. Fac Rev 2021; 10:21. [PMID: 33718938 PMCID: PMC7946390 DOI: 10.12703/r/10-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Organ transplantation is a life-saving treatment for end-stage organ failure. However, despite advances in immunosuppression, donor matching, tissue typing, and organ preservation, many organs are still lost each year to rejection. Ultimately, tolerance in the absence of immunosuppression is the goal, and although this seldom occurs spontaneously, a deeper understanding of alloimmunity may provide avenues for future therapies which aid in its establishment. Here, we highlight the recent key advances in our understanding of the allograft response. On the innate side, recent work has highlighted the previously unrecognised role of innate lymphoid cells as well as natural killer cells in promoting the alloresponse. The two major routes of allorecognition have recently been joined by a third newly identified pathway, semi-direct allorecognition, which is proving to be a key active pathway in transplantation. Through this review, we detail these newly defined areas in the allograft response and highlight areas for potential future therapeutic intervention.
Collapse
Affiliation(s)
- Conor Hennessy
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Guido Lewik
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Amy Cross
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
8
|
Pandolfi L, Fusco R, Frangipane V, D'Amico R, Giustra M, Bozzini S, Morosini M, D'Amato M, Cova E, Ferrario G, Morbini P, Colombo M, Prosperi D, Viglio S, Piloni D, Di Paola R, Cuzzocrea S, Meloni F. Loading Imatinib inside targeted nanoparticles to prevent Bronchiolitis Obliterans Syndrome. Sci Rep 2020; 10:20726. [PMID: 33244143 PMCID: PMC7693282 DOI: 10.1038/s41598-020-77828-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022] Open
Abstract
Bronchiolitis Obliterans Syndrome seriously reduces long-term survival of lung transplanted patients. Up to now there is no effective therapy once BOS is established. Nanomedicine introduces the possibility to administer drugs locally into lungs increasing drug accumulation in alveola reducing side effects. Imatinib was loaded in gold nanoparticles (GNP) functionalized with antibody against CD44 (GNP-HCIm). Lung fibroblasts (LFs) were derived from bronchoalveolar lavage of BOS patients. GNP-HCIm cytotoxicity was evaluated by MTT assay, apoptosis/necrosis and phosphorylated-cAbl (cAbl-p). Heterotopic tracheal transplantation (HTT) mouse model was used to evaluate the effect of local GNP-HCIm administration by Alzet pump. GNP-HCIm decreased LFs viability compared to Imatinib (44.4 ± 1.8% vs. 91.8 ± 3.2%, p < 0.001), inducing higher apoptosis (22.68 ± 4.3% vs. 6.43 ± 0.29; p < 0.001) and necrosis (18.65 ± 5.19%; p < 0.01). GNP-HCIm reduced cAbl-p (0.41 GNP-HCIm, 0.24 Imatinib vs. to control; p < 0.001). GNP-HCIm in HTT mouse model by Alzet pump significantly reduced tracheal lumen obliteration (p < 0.05), decreasing apoptosis (p < 0.05) and TGF-β-positive signal (p < 0.05) in surrounding tissue. GNP-HCIm treatment significantly reduced lymphocytic and neutrophil infiltration and mast cells degranulation (p < 0.05). Encapsulation of Imatinib into targeted nanoparticles could be considered a new option to inhibit the onset of allograft rejection acting on BOS specific features.
Collapse
Affiliation(s)
- Laura Pandolfi
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100, Pavia, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 981000, Messina, Italy
| | - Vanessa Frangipane
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100, Pavia, Italy
| | - Ramona D'Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 981000, Messina, Italy
| | - Marco Giustra
- NanoBioLab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20100, Milano, Italy
| | - Sara Bozzini
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100, Pavia, Italy
| | - Monica Morosini
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100, Pavia, Italy
| | - Maura D'Amato
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100, Pavia, Italy
| | - Emanuela Cova
- Department of Molecular Medicine, Pathology Unit, University of Pavia; IRCCS Foundation Policlinico San Matteo, 27100, Pavia, Italy
| | - Giuseppina Ferrario
- Department of Molecular Medicine, Pathology Unit, University of Pavia; IRCCS Foundation Policlinico San Matteo, 27100, Pavia, Italy
| | - Patrizia Morbini
- Department of Molecular Medicine, Pathology Unit, University of Pavia; IRCCS Foundation Policlinico San Matteo, 27100, Pavia, Italy
| | - Miriam Colombo
- NanoBioLab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20100, Milano, Italy
| | - Davide Prosperi
- NanoBioLab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20100, Milano, Italy.,Nanomedicine Laboratory, ICS Maugeri S.P.A., 27100, Pavia, Italy
| | - Simona Viglio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100, Pavia, Italy
| | - Davide Piloni
- Department of Internal Medicine, Section of Pneumology, University of Pavia, Pavia, Italy.,Department of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 981000, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 981000, Messina, Italy.,Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Federica Meloni
- Department of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation, Pavia, Italy.,Department of Internal Medicine, Section of Pneumology, University of Pavia, 27100, Pavia, Italy
| |
Collapse
|
9
|
LPS-induced Airway-centered Inflammation Leading to BOS-like Airway Remodeling Distinct From RAS-like Fibrosis in Rat Lung Transplantation. Transplantation 2020; 104:1150-1158. [PMID: 31929420 DOI: 10.1097/tp.0000000000003097] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Localization of inflammatory stimuli may direct lung allografts to different phenotypes of chronic dysfunction, such as bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS). We hypothesized that airway stimulation with lipopolysaccharide (LPS) in rats leads to airway-centered inflammation similar to human BOS. METHODS Rat left lung transplantation was conducted (donor: Brown Norway, recipient: Lewis). Allotransplant recipients received cyclosporine A (CsA) until postoperative day 56 with airway instillation of LPS (Allo-LPS, n = 8), phosphate buffered saline (Allo-PBS, n = 5) from days 35 to 46 (3 times a wk), or no further treatment (n = 4). Some allotransplant recipients received CsA until day 14 and were immunosuppression free after day 15 until day 56. Bronchial and pleural fibrosis were semiquantified; alveolar fibrosis was evaluated with a histological scale. RESULTS The Allo-LPS group had significantly increased International Society for Heart and Lung Transplantation rejection grades (grade A, P = 0.005; grade B, P = 0.004), bronchial obstructive proportion (0.34 ± 0.04% [Allo-LPS] versus 0.11 ± 0.04% [Allo-PBS], P = 0.006), and airway resistance (3.05 ± 1.78 cm H2O·s/mL [Allo-LPS] versus 0.83 ± 0.58 cm H2O·s/mL [Allo-PBS], P = 0.007) compared with other groups. Allotransplant recipients that underwent a short course of CsA developed RAS-like fibrosis involving the airways, alveoli, and pleura. CONCLUSIONS Airway instillation of LPS in allografts under immunosuppression resulted in BOS-like airway-centered inflammation and fibrosis distinct from RAS-like diffuse fibrosis, which was induced by a shortened course of immunosuppression. We propose novel animal models for BOS and RAS after lung transplantation.
Collapse
|
10
|
Dong M, Wang X, L T, Wang J, Yang Y, Liu Y, Jing Y, Zhao H, Chen J. Mir-27a-3p attenuates bronchiolitis obliterans in vivo via the regulation of dendritic cells' maturation and the suppression of myofibroblasts' differentiation. Clin Transl Med 2020; 10:e140. [PMID: 32898329 PMCID: PMC7423186 DOI: 10.1002/ctm2.140] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023] Open
Abstract
Bronchiolitis obliterans (BO), is a chronic rejection phenotype characterized by chronic small airway fibrous obliteration, hinders the patients who suffer from lung transplanting for surviving longer. Cell-based therapies using dendritic cells (DCs) and T regulatory cells (Tregs) have been developed to regulate allograft rejection, and to induce and maintain immune tolerance. In the present study, the effects of mir-27a-3p on regulating DCs as well as resulting effects on BO attenuation have been investigated. According to our reporter assays, the potential targets of mir-27a-3p were Smad2, sprouty2, and Smad4, respectively. Furthermore, sprouty2 inhibition by mir-27-3p indirectly activated extracellular regulated protein kinases (ERK) and increased IL-10 production in DCs. This led to a positive feedback loop that maintained the immature state of DCs via IL-10/JAK/STAT3 pathway, and caused an increase in Foxp3+ CD4+ T cells amount as well as TGF-β level. Furthermore, mir-27a-3p regulated TGF-β function, inhibited TGF-β/Smad pathway, and suppressed myofibroblast differentiation through influencing the function of Smad2 and Smad4. In short, the study indicated the effect of mir-27a-3p on suppressing DC maturation, which implicated the potential clinical application in treating postlung transplant BO.
Collapse
Affiliation(s)
- Ming Dong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Xin Wang
- Department of Pediatric Surgery, Tianjin Children's Hospital, Tianjin, P. R. China
| | - Tong L
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Jing Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Yunwei Yang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yi Liu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Yaqing Jing
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, P. R. China
| | - Honglin Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China
| |
Collapse
|
11
|
Li P, Zhu L, Tang FF, Xiong J, Ma MJ, Dsa M, Gao SH. A Simplified Continuous Two-stitch Suture for Bronchial Anastomosis of Left Single Lung Transplant in Dogs. Curr Med Sci 2020; 40:548-555. [PMID: 32681258 DOI: 10.1007/s11596-020-2212-2] [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: 11/27/2019] [Revised: 05/20/2020] [Indexed: 11/28/2022]
Abstract
Large animal models are essential to pre-clinical trials of pulmonary transplantation and bronchial anastomosis poses a great technical challenge to the procedure. Presented here is a simplified continuous two-stitch suture technique into bronchial anastomosis during the course of left single lung transplantation in canine. Animals were divided into three groups with each group having 6 animals. Left single lung transplantation in canine was performed to assess the feasibility of using this technique for bronchial anastomosis. In the control groups, all anastomoses were done by using traditional technique. Allograft functions and hemodynamic parameters were monitored during a 3-h reperfusion period. Quality of bronchial healing and airway complications were assessed by bronchoscopic surveillance after transplantation. We successfully completed left lung transplantation in 18 dogs, and all the dogs survived the procedures. The new technique substantially simplified the procedures for bronchial anastomosis and greatly reduced the time for bronchial anastomosis (P<<0.01) and the ischemic time of the grafts (P<0.05) compared to the control group. The continuous two-stitch suture attenuated the tissue injury to allografts and led to better blood gas exchange function as compared to the control group (P<0.05). Good bronchial healing (Grade I) was observed in all the groups. A canine left single lung transplantation model is feasible by using the novel suture technique, and the new technique is as safe as the traditional method. The technique is easy to learn, particularly for less experienced operators. Simpler and time-saving, the technique has great potential to be widely employed in clinical lung transplantation.
Collapse
Affiliation(s)
- Ping Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lan Zhu
- Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fei-Fei Tang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming-Jia Ma
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mouniir Dsa
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Si-Hai Gao
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory of Organ Transplantation, Chinese Ministry of Education and Chinese Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
12
|
Sato M. Bronchiolitis obliterans syndrome and restrictive allograft syndrome after lung transplantation: why are there two distinct forms of chronic lung allograft dysfunction? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:418. [PMID: 32355862 PMCID: PMC7186721 DOI: 10.21037/atm.2020.02.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bronchiolitis obliterans syndrome (BOS) had been considered to be the representative form of chronic rejection or chronic lung allograft dysfunction (CLAD) after lung transplantation. In BOS, small airways are affected by chronic inflammation and obliterative fibrosis, whereas peripheral lung tissue remains relatively intact. However, recognition of another form of CLAD involving multiple tissue compartments in the lung, termed restrictive allograft syndrome (RAS), raised a fundamental question: why there are two phenotypes of CLAD? Increasing clinical and experimental data suggest that RAS may be a prototype of chronic rejection after lung transplantation involving both cellular and antibody-mediated alloimmune responses. Some cases of RAS are also induced by fulminant general inflammation in lung allografts. However, BOS involves alloimmune responses and the airway-centered disease process can be explained by multiple mechanisms such as external alloimmune-independent stimuli (such as infection, aspiration and air pollution), exposure of airway-specific autoantigens and airway ischemia. Localization of immune responses in different anatomical compartments in different phenotypes of CLAD might be associated with lymphoid neogenesis or the de novo formation of lymphoid tissue in lung allografts. Better understanding of distinct mechanisms of BOS and RAS will facilitate the development of effective preventive and therapeutic strategies of CLAD.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
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: 5.8] [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
|
14
|
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: 2.8] [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
|
15
|
The prevention of tracheal graft occlusion using pioglitazone: A mouse tracheal transplant model study. Transpl Immunol 2019; 53:21-27. [DOI: 10.1016/j.trim.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022]
|
16
|
Kwak HH, Woo HM, Park KM. The degree of major histocompatibility complex matching between purebred Maltese and mongrel dogs using microsatellite markers. J Vet Sci 2019; 20:e5. [PMID: 30944528 PMCID: PMC6441805 DOI: 10.4142/jvs.2019.20.e5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/09/2019] [Accepted: 02/27/2019] [Indexed: 02/02/2023] Open
Abstract
Long-term maintenance of transplanted organs is one of the major factors that increases survival time of recipients. Although obtaining a major histocompatibility complex (MHC)-matched donor with the recipient is essential for successful organ transplantation, there have been limited reports on MHC matching between dogs. In this study, we analyzed the canine MHC matching rates using Maltese, one of the most popular purebred dogs, and mongrel dogs in Korea. Genomic DNA was extracted from blood leukocytes and DNA was amplified by polymerase chain reaction with primers specific to MHC microsatellite markers. The MHC matching degree was confirmed by the microsatellite markers using polyacrylamide gel electrophoresis. The MHC matching rates of each donor-recipient groups including Maltese-Maltese, mongrel-mongrel and Maltese-mongrel were 4.76%, 5.13% and 6.67%, respectively. There were no significant differences in the MHC matching degree between each group. These results demonstrate that MHC-matched donors could be selected from other breeds as much as from the same breed for transplantation. Knowledge of the MHC matching degree of purebred and mongrel dogs would offer valuable information not only for improving the success rate of organ transplantation surgery in canine patients but also for transplantation research using experimental canine models.
Collapse
Affiliation(s)
- Ho-Hyun Kwak
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Heung-Myong Woo
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Kyung-Mee Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
17
|
Takamori S, Shoji F, Okamoto T, Kozuma Y, Matsubara T, Haratake N, Akamine T, Katsura M, Takada K, Toyokawa G, Tagawa T, Maehara Y. HMGB1 blockade significantly improves luminal fibrous obliteration in a murine model of bronchiolitis obliterans syndrome. Transpl Immunol 2018; 53:13-20. [PMID: 30508580 DOI: 10.1016/j.trim.2018.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although high-mobility group box-1 (HMGB1), which is a nuclear protein, was reported to enhance the allogeneic responses in transplantation, the effect of HMGB1 on bronchiolitis obliterans syndrome (BOS) is unknown. METHODS A murine heterotopic tracheal transplantation model was used. Protein concentrations of HMGB1, interferon-γ (IFN-γ), interleukin (IL)-10, and IL-17 were analyzed in the isografts, allografts, controls, and HMGB1-neutralizing antibody administered allografts (n = 6; Days 1, 3, 5, 7, 14, 21, and 28). The luminal fibrous occlusion was analyzed (n = 6; Days 7, 14, 21, and 28). Infiltrating CD8 and CD4 T lymphocytes around the allografts and serum levels of IFN-γ and IL-10 were evaluated (n = 6; Day 7). RESULTS The HMGB1 levels in the allografts were significantly increased compared with the isografts at Day 7. HMGB1 blockade did not change the IL-17 level, but decreased the IFN-γ/IL-10 ratio in the early phase (Days 5 and 7) and significantly improved the fibrous occlusion in the late phase (Days 14, 21, and 28). HMGB1 blockade significantly suppressed the CD8 T lymphocytes infiltration and decreased the serum IFN-γ/IL-10 ratio compared with the control at Day 7. CONCLUSIONS HMGB1 may be a trigger of the BOS pathogenesis and candidate target for the treatment of the disease.
Collapse
Affiliation(s)
- Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Fumihiro Shoji
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan.
| | - Tatsuro Okamoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yuka Kozuma
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Taichi Matsubara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Naoki Haratake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Masakazu Katsura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Kazuki Takada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| |
Collapse
|
18
|
Yang M, Chen G, Zhang X, Ding Z, Miao Y, Yang Y, Chen ZK, Jiang F, Chang S, Zhou P. A novel MyD88 inhibitor attenuates allograft rejection after heterotopic tracheal transplantation in mice. Transpl Immunol 2018; 53:1-6. [PMID: 30472390 DOI: 10.1016/j.trim.2018.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 11/21/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND After lung transplantation, the major complication limiting the long-term survival of allografts is obliterative bronchiolitis (OB), characterized by chronic rejection. Innate immune responses contribute to the development of OB. In this study, we used a murine heterotopic tracheal transplantation mouse model to examine the effects of a newtype of innate immune inhibitor, TJ-M2010-5. METHODS Syngeneic tracheal grafts were transplanted heterotopically from C57BL/6 mice to C57BL/6 mice. Allografts from BALB/c mice were transplanted to C57BL/6 mice. The allograft recipients were treated with TJ-M2010-5, and anti-mouse CD154 (MR-1). The grafts were harvested at 7, 14, and 28 days and evaluated by histological and real-time RT-PCR analyses. RESULTS In untreated allografts, almost all epithelial cells fell off at 7 days and tracheal occlusion reached a peak at 28 days. However, the loss of the epithelium and airway obstruction were significantly improved in mice treated with TJ-M2010-5 combined with MR-1. The relative mRNA expression levels of pro-inflammatory cytokines were upregulated in allogeneic tracheal grafts, and treatment with the two drugs reduced the production of pro-inflammatory cytokines and infiltration of inflammatory cells. CONCLUSIONS In heterotopic tracheal transplantation models, TJ-M2010-5 combined with MR-1 could ameliorate the development of OB.
Collapse
Affiliation(s)
- Min Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Gen Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zuochuan Ding
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Yan Miao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Yang Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Zhonghua Klaus Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Fengchao Jiang
- Academy of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China.
| | - Ping Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China.
| |
Collapse
|
19
|
Kawashima M, Sato M, Murakawa T, Anraku M, Konoeda C, Hosoi A, Kakimi K, Nakajima J. Role of Toll-like Receptor 4 Expressed by Fibroblasts in Allograft Fibrosis in Mouse Orthotopic Tracheal Transplantation. Transplant Proc 2018; 50:3863-3872. [PMID: 30577279 DOI: 10.1016/j.transproceed.2018.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/25/2018] [Accepted: 06/21/2018] [Indexed: 12/16/2022]
Abstract
Development of chronic lung allograft dysfunction involves various alloimmune-independent insults including those mediated by Toll-like receptor (TLR) signaling, which is known to activate alloimmune responses. We hypothesized that TLR signaling may also contribute to the activation of fibroblasts and promoting allograft airway fibrosis. Mouse orthotopic tracheal transplants were conducted between major histocompatibility complex (MHC)-mismatched Balb/c donor and wild-type C3H or C3H-derived TLR4 mutant recipients (nonfunctional TLR4). Immunohistochemistry on day 21 showed significantly smaller alpha-smooth muscle actin (α-SMA)-positive areas in TLR4 mutant recipients than wild-type recipients (P = .01). No difference was found for CD3+ T-cell infiltration. Proliferation of alloreactive T cells derived from the recipient spleen showed no difference between TLR4 mutant and wild-type recipients in a mixed lymphocyte reaction. The effect of TLR4 signaling was examined in primary pulmonary fibroblast cultures both with lipopolysaccharide (LPS) and transforming growth factor (TGF)-β1. Stimulation with LPS significantly increased expression of α-SMA mRNA in wild-type fibroblasts cultured with TGF-β1 compared with the control without LPS (P = .001). Taken together, these findings suggest disruption of TLR signaling leads to reduced activation of fibroblasts without affecting T-cell infiltration and proliferation in this model. TLR4-mediated activation of fibroblasts may be a potentially important mechanism of allograft remodeling.
Collapse
Affiliation(s)
- M Kawashima
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - M Sato
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - T Murakawa
- Department of Thoracic Surgery, Kansai Medical University, Osaka, Japan
| | - M Anraku
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - C Konoeda
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - A Hosoi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - K Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - J Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Chang JC, Finn SM, Davis RP, Sanders NL, Holzknecht ZE, Everett ML, Barbas AS, Parker W, Lin SS. Early Immune Response to Acute Gastric Fluid Aspiration in a Rat Model of Lung Transplantation. EXP CLIN TRANSPLANT 2018; 17:84-92. [PMID: 29697356 DOI: 10.6002/ect.2017.0152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Chronic aspiration of gastric fluid contents can decrease long-term survival of pulmonary transplants due to development of obliterative bronchiolitis. However, little is known about the early immune response and the cascade of events involved in the development of obliterative bronchiolitis. MATERIALS AND METHODS We utilized a rat orthotopic pulmonary transplant model and a single aspiration of either gastric fluid or normal saline to investigate the histologic, cellular, and cytokine changes associated with an acute gastric fluid aspiration event compared with normal saline at 2 and 10 days after aspiration. RESULTS Our observations included a decrease in pulmonary compliance and increased airway inflammation and acute rejection of the transplanted lung, as well as increases in macrophages, granulocytes, and proinflammatory cytokines such as interleukin 1β, transforming growth factor β1 and β2, and tumor necrosis factor α in bronchoalveolar lavage fluid from the transplanted lung of gastric fluid-aspirated rats compared with normal saline-aspirated rats. CONCLUSIONS The acute inflammatory response observed in the present study is consistent with changes found in chronic models of aspiration-associated injury and suggests a potentially important role for mast cells in the development of obliterative bronchiolitis.
Collapse
Affiliation(s)
- Jui-Chih Chang
- From the Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA and the Department of Surgery, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Chronic Airway Fibrosis in Orthotopic Mouse Lung Transplantation Models—An Experimental Reappraisal? Transplantation 2018; 102:191-192. [DOI: 10.1097/tp.0000000000001998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Pentraxin 3 deficiency enhances features of chronic rejection in a mouse orthotopic lung transplantation model. Oncotarget 2018; 9:8489-8501. [PMID: 29492210 PMCID: PMC5823599 DOI: 10.18632/oncotarget.23902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/12/2017] [Indexed: 12/24/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is a serious complication after lung transplantation and thought to represent chronic rejection. Increased expression of Pentraxin 3 (PTX3), an acute phase protein, was associated with worse outcome in lung transplant patients. To determine the role of recipient PTX3 in development of chronic rejection, we used a minor alloantigen-mismatched murine orthotopic single lung transplant model. Male C57BL/10 mice were used as donors. Male PTX3 knockout (KO) mice and their wild type (WT) littermates on 129/SvEv/C57BL6/J background were used as recipients. In KO recipients, 7/13 grafted lungs were consolidated without volume recovery on CT scan, while only 2/9 WT mice showed similar graft consolidation. For grafts where lung volume could be reliably analyzed by CT scan, the lung volume recovery was significantly reduced in KO mice compared to WT. Interstitial inflammation, parenchymal fibrosis and bronchiolitis obliterans scores were significantly higher in KO mice. Presence of myofibroblasts and lymphoid aggregation was significantly enhanced in the grafts of PTX3 KO recipients. Recipient PTX3 deficiency enhanced chronic rejection-like lesions by promoting a fibrotic process in the airways and lung parenchyma. The underlying mechanisms and potential protective role of exogenous PTX3 as a therapy should be further explored.
Collapse
|
23
|
Kawakami T, Ito K, Matsuda Y, Noda M, Sakurada A, Hoshikawa Y, Okada Y, Ogasawara K. Cytotoxicity of Natural Killer Cells Activated Through NKG2D Contributes to the Development of Bronchiolitis Obliterans in a Murine Heterotopic Tracheal Transplant Model. Am J Transplant 2017; 17:2338-2349. [PMID: 28251796 DOI: 10.1111/ajt.14257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 01/25/2023]
Abstract
Bronchiolitis obliterans after lung transplantation is a major cause of postoperative mortality in which T cell-mediated immunity is known to play an important role. However, the exact contribution of natural killer (NK) cells, which have functions similar to CD8+ T cells, has not been defined. Here, we assessed the role of NK cells in murine bronchiolitis obliterans through heterotopic tracheal transplantations and found a greater percentage of NK cells in allografts than in isografts. Depletion of NK cells using an anti-NK1.1 antibody attenuated bronchiolitis obliterans in transplant recipients compared with controls. In terms of NK cell effector functions, an improvement in bronchiolitis obliterans was observed in perforin-KO recipient mice compared to wild type (WT). Furthermore, we found upregulation of NKG2D-ligand in allografts and demonstrated the significance of this using grafts expressing Rae-1, a murine NKG2D-ligand, which induced severe bronchiolitis obliterans in WT and Rag-1 KO recipients. This effect was ameliorated by injection of anti-NKG2D blocking antibody. Together, these results suggest that cytotoxicity resulting from activation of NK cells through NKG2D leads to the development of murine bronchiolitis obliterans.
Collapse
Affiliation(s)
- T Kawakami
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan.,Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - K Ito
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Y Matsuda
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - M Noda
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - A Sakurada
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Y Hoshikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Y Okada
- Department of Thoracic Surgery, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - K Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
24
|
Miyamoto E, Motoyama H, Sato M, Aoyama A, Menju T, Shikuma K, Sowa T, Yoshizawa A, Saito M, Takahagi A, Tanaka S, Takahashi M, Ohata K, Kondo T, Hijiya K, Chen-Yoshikawa TF, Date H. Association of Local Intrapulmonary Production of Antibodies Specific to Donor Major Histocompatibility Complex Class I With the Progression of Chronic Rejection of Lung Allografts. Transplantation 2017; 101:e156-e165. [PMID: 28207638 DOI: 10.1097/tp.0000000000001665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Antibody-mediated rejection may lead to chronic lung allograft dysfunction, but antibody-mediated rejection may develop in the absence of detectable donor-specific antibody (DSA) in recipient serum. This study investigated whether humoral immune responses develop not only systemically but locally within rejected lung allografts, resulting in local production of DSA. METHODS Lewis rats received orthotopic left lung transplantation from Lewis (syngeneic control) or Brown-Norway (major histocompatibility complex-mismatched allogeneic) donor rats. Rats that underwent allogeneic lung transplantation were subsequently administered cyclosporine until day 14 (short immunosuppression) or day 35 (long immunosuppression). The lung grafts and spleens of recipient animals were tissue cultured for 4 days, and the titer of antibody against donor major histocompatibility complex molecules was assayed by flow cytometry. Explanted lung grafts were also evaluated pathologically. RESULTS By day 98, DSA titers in supernatants of lung graft (P = 0.0074) and spleen (P = 0.0167) cultures, but not serum, from the short immunosuppression group were significantly higher than titers in syngeneic controls. Cultures and sera from the long immunosuppression group showed no production of DSA. Microscopically, the lung grafts from the short immunosuppression group showed severe bronchiole obliteration and parenchymal fibrosis, along with lymphoid aggregates containing T and B cells, accompanying plasma cells. These findings suggestive of local humoral immune response were not observed by days 28 and 63. CONCLUSIONS DSA can be locally produced in chronically rejected lung allografts, along with intragraft immunocompetent cells. Clinical testing of DSA in serum samples alone may underestimate lung allograft dysfunction.
Collapse
Affiliation(s)
- Ei Miyamoto
- 1 Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan. 2 Department of Thoracic Surgery, The University of Tokyo Hospital, Tokyo, Japan. 3 Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lorenzi W, Gonçalves FDC, Schneider N, Silva ÉF, Visioli F, Paz AH, Saueressig MG. Repeated systemic administration of adipose tissue-derived mesenchymal stem cells prevents tracheal obliteration in a murine model of bronchiolitis obliterans. Biotechnol Lett 2017; 39:1269-1277. [PMID: 28528427 DOI: 10.1007/s10529-017-2355-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/04/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate the effect of adipose tissue-derived mesenchymal stem cell (ASC) administered either systemically or locally in a murine model of bronchiolitis obliterans. RESULTS When compared to controls, systemic treatment with 106 ASCs on D0 and a second dose on D7 significantly prevented tracheal obliteration 28 days after heterotopic tracheal transplantation (median of 94 vs. 16%; P < 0.01). A single dose tended towards less stenosis than controls, but did not reach statistical significance (28 vs. 94%; P = 0.054). On the contrary, repeated local injection was incapable of preventing tracheal obliteration when compared to a single injection or controls (37 vs. 71 vs. 87%). Two intravenous doses also tended to be better than two local injections (16 vs. 37%; P = 0.058), and were better than a single local dose (16 vs. 71%; P < 0.01). CONCLUSION A second dose of ASC, given systemically after 7 days, reduces luminal obliteration in a heterotopic tracheal transplantation model in mice, suggesting that ASC can be used to prevent obliterative bronchiolitis after lung transplantation.
Collapse
Affiliation(s)
- William Lorenzi
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil.
| | - Fabiany Da Costa Gonçalves
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Natália Schneider
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Éverton Franco Silva
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Fernanda Visioli
- Pathology Department, School of Odontology, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2492, Porto Alegre, RS, CEP 90035-007, Brazil
| | - Ana Helena Paz
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mauricio Guidi Saueressig
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil
| |
Collapse
|
26
|
Lama VN, Belperio JA, Christie JD, El-Chemaly S, Fishbein MC, Gelman AE, Hancock WW, Keshavjee S, Kreisel D, Laubach VE, Looney MR, McDyer JF, Mohanakumar T, Shilling RA, Panoskaltsis-Mortari A, Wilkes DS, Eu JP, Nicolls MR. Models of Lung Transplant Research: a consensus statement from the National Heart, Lung, and Blood Institute workshop. JCI Insight 2017; 2:93121. [PMID: 28469087 DOI: 10.1172/jci.insight.93121] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lung transplantation, a cure for a number of end-stage lung diseases, continues to have the worst long-term outcomes when compared with other solid organ transplants. Preclinical modeling of the most common and serious lung transplantation complications are essential to better understand and mitigate the pathophysiological processes that lead to these complications. Various animal and in vitro models of lung transplant complications now exist and each of these models has unique strengths. However, significant issues, such as the required technical expertise as well as the robustness and clinical usefulness of these models, remain to be overcome or clarified. The National Heart, Lung, and Blood Institute (NHLBI) convened a workshop in March 2016 to review the state of preclinical science addressing the three most important complications of lung transplantation: primary graft dysfunction (PGD), acute rejection (AR), and chronic lung allograft dysfunction (CLAD). In addition, the participants of the workshop were tasked to make consensus recommendations on the best use of these complimentary models to close our knowledge gaps in PGD, AR, and CLAD. Their reviews and recommendations are summarized in this report. Furthermore, the participants outlined opportunities to collaborate and directions to accelerate research using these preclinical models.
Collapse
Affiliation(s)
- Vibha N Lama
- Department of Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jason D Christie
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Souheil El-Chemaly
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael C Fishbein
- Department of Pathology and Laboratory Medicine, UCLA Center for the Health Sciences, Los Angeles, California, USA
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wayne W Hancock
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shaf Keshavjee
- Division of Thoracic Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark R Looney
- Department of Medicine, UCSF School of Medicine, San Francisco, California, USA
| | - John F McDyer
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Rebecca A Shilling
- Department of Medicine, University of Illinois College of Medicine at Chicago, Illinois, USA
| | - Angela Panoskaltsis-Mortari
- Departments of Pediatrics, and Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - David S Wilkes
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jerry P Eu
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mark R Nicolls
- Department of Medicine, Stanford University School of Medicine/VA Palo Alto Health Care System, Stanford, California, USA
| |
Collapse
|
27
|
Apoptosis of the Tracheal Epithelium Can Increase the Number of Recipient Bone Marrow–Derived Myofibroblasts in Allografts and Exacerbate Obliterative Bronchiolitis After Tracheal Transplantation in Mice. Transplantation 2016; 100:1880-8. [DOI: 10.1097/tp.0000000000001230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Verleden SE, Sacreas A, Vos R, Vanaudenaerde BM, Verleden GM. Advances in Understanding Bronchiolitis Obliterans After Lung Transplantation. Chest 2016; 150:219-25. [PMID: 27212132 DOI: 10.1016/j.chest.2016.04.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 03/29/2016] [Accepted: 04/13/2016] [Indexed: 11/30/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) remains a major complication after lung transplantation, causing significant morbidity and mortality in a majority of recipients. BOS is believed to be the clinical correlate of chronic allograft dysfunction, and is defined as an obstructive pulmonary function defect in the absence of other identifiable causes, mostly not amenable to treatment. Recently, it has become clear that BOS is not the only form of chronic allograft dysfunction and that other clinical phenotypes exist; however, we focus exclusively on BOS. Radiologic findings typically demonstrate air trapping, mosaic attenuation, and hyperinflation. Pathologic examination reveals obliterative bronchiolitis lesions and a pure obliteration of the small airways (< 2 mm), with a relatively normal surrounding parenchyma. In this review, we highlight recent advances in diagnosis, pathologic examination, and risk factors, such as microbes, viruses, and antibodies. Although the pathophysiological mechanisms remain largely unknown, we review the role of the airway epithelium and inflammation and the various experimental animal models. We also clarify the clinical and therapeutic implications of these findings. Although significant progress has been made, the exact pathophysiological mechanisms and adequate therapy for posttransplantation BOS remain unknown, highlighting the need for further research to improve long-term posttransplantation BOS-free and overall survival.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Ropponen JO, Keränen MA, Raissadati A, Nykänen AI, Krebs R, Lemström KB, Tikkanen JM. Increased myeloid cell hypoxia-inducible factor-1 delays obliterative airway disease in the mouse. J Heart Lung Transplant 2016; 35:671-8. [DOI: 10.1016/j.healun.2015.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 11/05/2015] [Accepted: 12/15/2015] [Indexed: 11/26/2022] Open
|
30
|
Halofuginone treatment reduces interleukin-17A and ameliorates features of chronic lung allograft dysfunction in a mouse orthotopic lung transplant model. J Heart Lung Transplant 2016; 35:518-27. [DOI: 10.1016/j.healun.2015.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/17/2015] [Accepted: 12/04/2015] [Indexed: 11/23/2022] Open
|
31
|
Nemska S, Daubeuf F, Frossard N. Revascularization of the graft in obliterative bronchiolitis after heterotopic tracheal transplantation. Physiol Rep 2016; 4:e12690. [PMID: 26908711 PMCID: PMC4816893 DOI: 10.14814/phy2.12690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 01/12/2023] Open
Abstract
Obliterative bronchiolitis is the principal long-term problem for lung transplant patients. One of the simplest and most reproducible animal models of obliterative bronchiolitis is heterotopic tracheal transplantation in subcutaneous tissue, where the graft is not primarily vascularized. We demonstrate here the rapid graft revascularization and the kinetics of expression of its angiogenic and lymphatic factors. We performed iso- and allotracheal transplantations harvested on day 0-21. The number of functional blood vessels, quantified after intravenous biotinylated dextran administration, increased from D0 (0 for both iso- and allografts) to D21 (44 ± 8 vessels/mm(2) in isografts and 22 ± 3 in allografts, P < 0.001 for both vs. D0). VEGF mRNA expression assessed by qPCR peaked on D1 (4.3-fold increase in isografts and 4.0-fold in allografts, P < 0.0001 for both vs. D0), but receded thereafter. Angiopoietin-1, involved in the maturation of the neoformed vessels, increased later on, by 6.2-fold (P < 0.05) in isografts and 11.5-fold in allografts (P < 0.001) by D21, and angiopoietin-2 by 7.8-fold in isografts (P < 0.05) and 13.8-fold in allografts (P < 0.01). Although always present in the iso- and allografts, there were significantly more and larger LYVE1(+) lymphatic vessels at D21 in allografts than in isografts. Thus, we demonstrate that tracheal grafts are rapidly revascularized by functional blood and lymphatic vessels, due to early VEGF and subsequent angiopoietins expression, which is a new advantage of this model, in addition to its ease of use, reproducibility, and viability in the absence of immunosuppressive treatment.
Collapse
Affiliation(s)
- Simona Nemska
- Laboratoire d'Innovation Thérapeutique, Unité Mixte de Recherche 7200 Centre National de la Recherche Scientifique-Université de Strasbourg and Laboratoire d'Excellence MEDALIS Faculté de Pharmacie, Illkirch, France
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique, Unité Mixte de Recherche 7200 Centre National de la Recherche Scientifique-Université de Strasbourg and Laboratoire d'Excellence MEDALIS Faculté de Pharmacie, Illkirch, France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Unité Mixte de Recherche 7200 Centre National de la Recherche Scientifique-Université de Strasbourg and Laboratoire d'Excellence MEDALIS Faculté de Pharmacie, Illkirch, France
| |
Collapse
|
32
|
Zhao Y, Gillen JR, Meher AK, Burns JA, Kron IL, Lau CL. Rapamycin prevents bronchiolitis obliterans through increasing infiltration of regulatory B cells in a murine tracheal transplantation model. J Thorac Cardiovasc Surg 2016; 151:487-96.e3. [PMID: 26481278 PMCID: PMC4728002 DOI: 10.1016/j.jtcvs.2015.08.116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/06/2015] [Accepted: 08/11/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE B lymphocytes are generally considered to be activators of the immune response; however, recent findings have shown that a subtype of B lymphocytes, regulatory B lymphocytes, play a role in attenuating the immune response. Bronchiolitis obliterans remains the major limitation to modern-day lung transplantation. The role of regulatory B lymphocytes in bronchiolitis obliterans has not been elucidated. We hypothesized that regulatory B lymphocytes play a role in the attenuation of bronchiolitis obliterans. METHODS We performed a standard heterotopic tracheal transplant model. Tracheas from Balb/c mice were transplanted into C57BL/6 recipients. Rapamycin treatment and dimethyl sulfoxide control groups were each treated for the first 14 days after the transplant. Tracheas were collected on days 7, 14, and 28 post-transplantation. Luminal obliteration was evaluated by hematoxylin-eosin staining and Picrosirius red staining. Immune cell infiltration and characteristics, and secretion of interleukin-10 and transforming growth factor-β1 were accessed by immunohistochemistry. Cytokines and transforming growth factor-β1 were measured using the Luminex assay (Bio-Rad, Hercules, Calif). RESULTS The results revealed that intraperitoneal injection of rapamycin for 14 days after tracheal transplantation significantly reduced luminal obliteration on day 28 when compared with the dimethyl sulfoxide control group (97.78% ± 3.63% vs 3.02% ± 2.14%, P < .001). Rapamycin treatment markedly induced regulatory B lymphocytes (B220(+)IgM(+)IgG(-)IL-10(+)TGF-β1(+)) cells when compared with dimethyl sulfoxide controls. Rapamycin treatment inhibited interleukin-1β, 6, 13, and 17 on days 7 and 14. Rapamycin also greatly increased interleukin-10 and transforming growth factor-β1 production in B cells and regulatory T lymphocytes infiltration on day 28. CONCLUSIONS Mammalian target of rapamycin inhibition decreases the development of bronchiolitis obliterans via inhibition of proinflammatory cytokines and increasing regulatory B lymphocytes cell infiltration, which subsequently produces anti-inflammatory cytokines and upregulates regulatory T lymphocyte cells.
Collapse
Affiliation(s)
- Yunge Zhao
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Jacob R Gillen
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Akshaya K Meher
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Jordan A Burns
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Irving L Kron
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Christine L Lau
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va.
| |
Collapse
|
33
|
Adams AB, Kitchens WH, Newell KA. Experimental models in discovery and translational studies. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Jiang X, Nguyen TT, Tian W, Sung YK, Yuan K, Qian J, Rajadas J, Sallenave JM, Nickel NP, de Jesus Perez V, Rabinovitch M, Nicolls MR. Cyclosporine Does Not Prevent Microvascular Loss in Transplantation but Can Synergize With a Neutrophil Elastase Inhibitor, Elafin, to Maintain Graft Perfusion During Acute Rejection. Am J Transplant 2015; 15:1768-81. [PMID: 25727073 PMCID: PMC4474772 DOI: 10.1111/ajt.13189] [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: 09/10/2014] [Revised: 12/10/2014] [Accepted: 12/26/2014] [Indexed: 01/25/2023]
Abstract
The loss of a functional microvascular bed in rejecting solid organ transplants is correlated with fibrotic remodeling and chronic rejection; in lung allografts, this pathology is predicted by bronchoalveolar fluid neutrophilia which suggests a role for polymorphonuclear cells in microcirculatory injury. In a mouse orthotopic tracheal transplant model, cyclosporine, which primarily inhibits T cells, failed as a monotherapy for preventing microvessel rejection and graft ischemia. To target neutrophil action that may be contributing to vascular injury, we examined the effect of a neutrophil elastase inhibitor, elafin, on the microvascular health of transplant tissue. We showed that elafin monotherapy prolonged microvascular perfusion and enhanced tissue oxygenation while diminishing the infiltration of neutrophils and macrophages and decreasing tissue deposition of complement C3 and the membrane attack complex, C5b-9. Elafin was also found to promote angiogenesis through activation of the extracellular signal-regulated kinase (ERK) signaling pathway but was insufficient as a single agent to completely prevent tissue ischemia during acute rejection episodes. However, when combined with cyclosporine, elafin effectively preserved airway microvascular perfusion and oxygenation. The therapeutic strategy of targeting neutrophil elastase activity alongside standard immunosuppression during acute rejection episodes may be an effective approach for preventing the development of irreversible fibrotic remodeling.
Collapse
Affiliation(s)
- Xinguo Jiang
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Tom T. Nguyen
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Wen Tian
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Yon K. Sung
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ke Yuan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jin Qian
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Jean-Michel Sallenave
- Unité de Défense Innée et Inflammation, Institut Pasteur, Paris, France,INSERM U884, Paris, France
| | - Nils P. Nickel
- Cardiovascular Institute and Department of Pediatrics, Stanford, CA
| | - Vinicio de Jesus Perez
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Mark R. Nicolls
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
35
|
Allogeneic CD4+CD25high T cells regulate obliterative bronchiolitis of heterotopic bronchus allografts in both porcinized and humanized mouse models. Transplantation 2015; 99:482-91. [PMID: 25695787 DOI: 10.1097/tp.0000000000000632] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bronchiolitis obliterans syndrome is caused by a fibroproliferative process in lung allografts resulting in irreversible damage. In this study, we induced obliterative bronchiolitis and studied the contribution of regulatory T cells to its development in immune-deficient mice receiving heterotopic porcine bronchus transplants, and major histocompatibility complex-mismatched porcine peripheral blood mononuclear cell. Furthermore, we aimed to corroborate our findings in a humanized mouse model. METHODS Heterotopic bronchus transplantation was performed in 33 NOD.rag(−/−)γc(−/−) mice, using miniature pigs as tissue donors.The recipient mice then either received saline (negative control), unsorted MHC-mismatched PBMC (positive control), PBMC enriched with CD4(+)CD25(high) cells or PBMC depleted of CD4(+)CD25(high) cells for reconstitution. The results were validated in 28 NOD.rag(−/−)γc(−/−) mice undergoing heterotopic human bronchus transplantation and reconstitution with allogeneic human PBMC. RESULTS Histological lesions similar to those typical for obliterative bronchiolitis developed in vivo after reconstitution with allogeneic PBMC and were more severe in animals engrafted with PBMC depleted of CD4(+)CD25(high) cells. In contrast, the group reconstituted with PBMC enriched with CD4(+)CD25(high) cells showed well-preserved histology. The results of the humanized model confirmed those obtained in the porcinized model. CONCLUSIONS In conclusion, both porcinized and humanized mouse models of heterotopic subcutaneous bronchus transplantation imitate the in vivo development of bronchiolitis obliterans syndrome-like lesions and reveal its sensitivity to T-cell regulation.
Collapse
|
36
|
Mimura T, Walker N, Aoki Y, Manning CM, Murdock BJ, Myers JL, Lagstein A, Osterholzer JJ, Lama VN. Local origin of mesenchymal cells in a murine orthotopic lung transplantation model of bronchiolitis obliterans. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1564-74. [PMID: 25848843 DOI: 10.1016/j.ajpath.2015.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 10/23/2022]
Abstract
Bronchiolitis obliterans is the leading cause of chronic graft failure and long-term mortality in lung transplant recipients. Here, we used a novel murine model to characterize allograft fibrogenesis within a whole-lung microenvironment. Unilateral left lung transplantation was performed in mice across varying degrees of major histocompatibility complex mismatch combinations. B6D2F1/J (a cross between C57BL/6J and DBA/2J) (Haplotype H2b/d) lungs transplanted into DBA/2J (H2d) recipients were identified to show histopathology for bronchiolitis obliterans in all allogeneic grafts. Time course analysis showed an evolution from immune cell infiltration of the bronchioles and vessels at day 14, consistent with acute rejection and lymphocytic bronchitis, to subepithelial and intraluminal fibrotic lesions of bronchiolitis obliterans by day 28. Allografts at day 28 showed a significantly higher hydroxyproline content than the isografts (33.21 ± 1.89 versus 22.36 ± 2.33 μg/mL). At day 40 the hydroxyproline content had increased further (48.91 ± 7.09 μg/mL). Flow cytometric analysis was used to investigate the origin of mesenchymal cells in fibrotic allografts. Collagen I-positive cells (89.43% ± 6.53%) in day 28 allografts were H2Db positive, showing their donor origin. This novel murine model shows consistent and reproducible allograft fibrogenesis in the context of single-lung transplantation and represents a major step forward in investigating mechanisms of chronic graft failure.
Collapse
Affiliation(s)
- Takeshi Mimura
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Natalie Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Yoshiro Aoki
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Casey M Manning
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Benjamin J Murdock
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Jeffery L Myers
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan
| | - Amir Lagstein
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan
| | - John J Osterholzer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Vibha N Lama
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan.
| |
Collapse
|
37
|
Morgan DL, Merrick BA, Gerrish KE, Stockton PS, Wang Y, Foley JF, Gwinn WM, Kelly FL, Palmer SM, Ton TVT, Flake GP. Gene expression in obliterative bronchiolitis-like lesions in 2,3-pentanedione-exposed rats. PLoS One 2015; 10:e0118459. [PMID: 25710175 PMCID: PMC4339611 DOI: 10.1371/journal.pone.0118459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 01/19/2015] [Indexed: 11/21/2022] Open
Abstract
Obliterative bronchiolitis (OB) is an irreversible lung disease characterized by progressive fibrosis in the small airways with eventual occlusion of the airway lumens. OB is most commonly associated with lung transplant rejection; however, OB has also been diagnosed in workers exposed to artificial butter flavoring (ABF) vapors. Research has been limited by the lack of an adequate animal model of OB, and as a result the mechanism(s) is unclear and there are no effective treatments for this condition. Exposure of rats to the ABF component, 2,3-pentanedione (PD) results in airway lesions that are histopathologically similar to those in human OB. We used this animal model to evaluate changes in gene expression in the distal bronchi of rats with PD-induced OB. Male Wistar Han rats were exposed to 200 ppm PD or air 6 h/d, 5 d/wk for 2-wks. Bronchial tissues were laser microdissected from serial sections of frozen lung. In exposed lungs, both fibrotic and non-fibrotic airways were collected. Following RNA extraction and microarray analysis, differential gene expression was evaluated. In non-fibrotic bronchi of exposed rats, 4683 genes were significantly altered relative to air-exposed controls with notable down-regulation of many inflammatory cytokines and chemokines. In contrast, in fibrotic bronchi, 3807 genes were significantly altered with a majority of genes being up-regulated in affected pathways. Tgf-β2 and downstream genes implicated in fibrosis were significantly up-regulated in fibrotic lesions. Genes for collagens and extracellular matrix proteins were highly up-regulated. In addition, expression of genes for peptidases and peptidase inhibitors were significantly altered, indicative of the tissue remodeling that occurs during airway fibrosis. Our data provide new insights into the molecular mechanisms of OB. This new information is of potential significance with regard to future therapeutic targets for treatment.
Collapse
Affiliation(s)
- Daniel L. Morgan
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| | - B. Alex Merrick
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Kevin E. Gerrish
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Patricia S. Stockton
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Yu Wang
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Julie F. Foley
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - William M. Gwinn
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Francine L. Kelly
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Scott M. Palmer
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Thai-Vu T. Ton
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Gordon P. Flake
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
38
|
Zhao J, Wang Y, Wakeham A, Hao Z, Toba H, Bai X, Keshavjee S, Mak TW, Liu M. XB130 deficiency affects tracheal epithelial differentiation during airway repair. PLoS One 2014; 9:e108952. [PMID: 25272040 PMCID: PMC4182764 DOI: 10.1371/journal.pone.0108952] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 08/26/2014] [Indexed: 12/26/2022] Open
Abstract
The repair and regeneration of airway epithelium is important for maintaining homeostasis of the respiratory system. XB130 is an adaptor protein involved in the regulation of cell proliferation, survival and migration. In the human trachea, XB130 is expressed on the apical site of ciliated epithelial cells. We hypothesize that XB130 may play a role in epithelial repair and regeneration after injury. Xb130 knockout (KO) mice were generated, and a mouse isogenic tracheal transplantation model was used. Adult Xb130 KO mice did not show any significant anatomical and physiological phenotypes in comparison with their wild type (WT) littermates. The tracheal epithelium in Xb130 KO mice, however, was significantly thicker than that in WT mice. Severe ischemic epithelial injury was observed immediately after the tracheal transplantation, which was followed by epithelial cell flattening, proliferation and differentiation. No significant differences were observed in terms of initial airway injury and apoptosis. However, at Day 10 after transplantation, the epithelial layer was significantly thicker in Xb130 KO mice, and associated with greater proliferative (Ki67+) and basal (CK5+) cells, as well as thickening of the connective tissue and fibroblast layer between the epithelium and tracheal cartilages. These results suggest that XB130 is involved in the regulation of airway epithelial differentiation, especially during airway repair after injury.
Collapse
Affiliation(s)
- Jinbo Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Thoracic Surgery, Tangdu Hospital, Forth Military Medical University, Xi’an, Shaanxi, China
| | - Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Andrew Wakeham
- Advanced Medical Discovery Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhenyue Hao
- Advanced Medical Discovery Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hiroaki Toba
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Xiaohui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tak W. Mak
- Advanced Medical Discovery Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
39
|
O'Koren EG, Hogan BLM, Gunn MD. Loss of basal cells precedes bronchiolitis obliterans-like pathological changes in a murine model of chlorine gas inhalation. Am J Respir Cell Mol Biol 2013; 49:788-97. [PMID: 23742075 DOI: 10.1165/rcmb.2012-0369oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchiolitis obliterans (BO) is a major cause of chronic airway dysfunction after toxic chemical inhalation. The pathophysiology of BO is not well understood, but epithelial cell injury has been closely associated with the development of fibrotic lesions in human studies and in animal models of both toxin-induced and transplant-induced BO. However, whereas almost all cases and models of BO include epithelial injury, not all instances of epithelial injury result in BO, suggesting that epithelial damage per se is not the critical event leading to the development of BO. Here, we describe a model of chlorine-induced BO in which mice develop tracheal and large airway obliterative lesions within 10 days of exposure to high (350 parts per million [ppm]), but not low (200 ppm), concentrations of chlorine gas. Importantly, these lesions arise only under conditions and in areas in which basal cells, the resident progenitor cells for large airway epithelium, are eliminated by chlorine exposure. In areas of basal cell loss, epithelial regeneration does not occur, resulting in persistent regions of epithelial denudation. Obliterative airway lesions arise specifically from regions of epithelial denudation in a process that includes inflammatory cell infiltration by Day 2 after exposure, fibroblast infiltration and collagen deposition by Day 5, and the ingrowth of blood vessels by Day 7, ultimately leading to lethal airway obstruction by Days 9-12. We conclude that the loss of epithelial progenitor cells constitutes a critical factor leading to the development of obliterative airway lesions after chemical inhalation.
Collapse
|
40
|
Preidl RHM, Eckl S, Ramsperger-Gleixner M, Koch N, Spriewald BM, Weyand M, Ensminger SM. Clopidogrel reduces post-transplant obliterative bronchiolitis. Transpl Int 2013; 26:1038-48. [DOI: 10.1111/tri.12163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/13/2013] [Accepted: 07/15/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Raimund H. M. Preidl
- Department of Cardiac Surgery; Friedrich-Alexander University; Erlangen-Nürnberg Germany
| | - Sebastian Eckl
- Department of Cardiac Surgery; Friedrich-Alexander University; Erlangen-Nürnberg Germany
| | | | - Nina Koch
- Department of Cardiac Surgery; Friedrich-Alexander University; Erlangen-Nürnberg Germany
| | - Bernd M. Spriewald
- Department of Internal Medicine 5; Hematology/Oncology and Institute of Clinical Immunology; 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
| |
Collapse
|
41
|
Fan K, Qiao XW, Nie J, Yuan L, Guo HZ, Zheng ZK, Li JS, Wang JJ, Jiang K. Orthotopic and heterotopic tracheal transplantation model in studying obliterative bronchiolitis. Transpl Immunol 2013; 28:170-5. [DOI: 10.1016/j.trim.2013.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/12/2013] [Accepted: 04/15/2013] [Indexed: 12/21/2022]
|
42
|
Subramanian V, Mohanakumar T. Chronic rejection: a significant role for Th17-mediated autoimmune responses to self-antigens. Expert Rev Clin Immunol 2013; 8:663-72. [PMID: 23078063 DOI: 10.1586/eci.12.58] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite progress in the field of organ transplantation for improvement in graft survival and function, long-term graft function is still limited by the development of chronic allograft rejection. Various immune-mediated and nonimmune-mediated processes have been postulated in the pathogenesis of chronic rejection. In this review, the authors discuss the important role of alloimmune responses to donor-specific antigens and autoimmune responses to tissue restricted self-antigens in the immunopathogenesis of chronic rejection following solid organ transplantation. In particular, the authors discuss the role of induction of Th17-type autoimmune responses and the crosstalk between autoimmune and alloimmune responses. These self-perpetuate each other leading to activation of profibrotic and proinflammatory cascades that ultimately result in the development of chronic rejection.
Collapse
Affiliation(s)
- Vijay Subramanian
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
43
|
Guo Z, Zhou X, Li J, Meng Q, Cao H, Kang L, Ni Y, Fan H, Liu Z. Mesenchymal stem cells reprogram host macrophages to attenuate obliterative bronchiolitis in murine orthotopic tracheal transplantation. Int Immunopharmacol 2013; 15:726-34. [DOI: 10.1016/j.intimp.2013.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/04/2013] [Accepted: 03/01/2013] [Indexed: 02/06/2023]
|
44
|
Densmore JC, Jeziorczak PM, Clough AV, Pritchard KA, Cummens B, Medhora M, Rao A, Jacobs ER. Rattus model utilizing selective pulmonary ischemia induces bronchiolitis obliterans organizing pneumonia. Shock 2013; 39:271-7. [PMID: 23364425 PMCID: PMC3578046 DOI: 10.1097/shk.0b013e318281a58c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Bronchiolitis obliterans organizing pneumonia (BOOP), a morbid condition when associated with lung transplant and chronic lung disease, is believed to be a complication of ischemia. Our goal was to develop a simple and reliable model of lung ischemia in the Sprague-Dawley rat that would produce BOOP. Unilateral ischemia without airway occlusion was produced by an occlusive slipknot placed around the left main pulmonary artery. Studies were performed 7 days later. Relative pulmonary and systemic flow to each lung was measured by injection of technetium Tc 99m macroaggregated albumin. Histological sections were examined for structure and necrosis and scored for BOOP. Apoptosis was detected by immunohistochemistry with an antibody against cleaved caspase 3. Pulmonary artery blood flow to left lungs was less than 0.1% of the cardiac output, and bronchial artery circulation was ∼2% of aortic artery flow. Histological sections from ischemic left lungs consistently showed Masson bodies, inflammation, and young fibroblasts filling the distal airways and alveoli, consistent with BOOP. In quantitative evaluation of BOOP using epithelial changes, inflammation and fibrosis were higher in ischemic left lungs than right or sham-operated left lungs. Apoptosis was increased in areas exhibiting histological BOOP, but there was no histological evidence of necrosis. Toll-like receptor 4 expression was increased in ischemic left lungs over right. An occlusive slipknot around the main left pulmonary artery in rats produces BOOP, providing direct evidence that ischemia without immunomodulation or coinfection is sufficient to initiate this injury. It also affords an excellent model to study signaling and genetic mechanisms underlying BOOP.
Collapse
|
45
|
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: 22] [Impact Index Per Article: 2.0] [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
|
46
|
Guo H, Nie J, Fan K, Zheng Z, Qiao X, Li J, Wang J, Jiang K. Improvements of surgical techniques in a rat model of an orthotopic single lung transplant. Eur J Med Res 2013; 18:1. [PMID: 23295132 PMCID: PMC3608166 DOI: 10.1186/2047-783x-18-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 12/07/2012] [Indexed: 12/15/2022] Open
Abstract
Background Rats are widely used in modeling orthotopic lung transplantation. Recently the introduction of the cuff technique has greatly facilitated the anastomosing procedure used during the transplant. However, the procedure is still associated with several drawbacks including twisting of blood vessels, tissue injury and the extensive time required for the procedure. This study was performed to optimize the model of rat lung transplantation (LT) with the cuff technique. Methods A total of 42 adult Lewis rats received orthotopic LT with our newly modified procedures. The modified procedures were based on the traditional procedure and incorporated improvements involving orotracheal intubation; a cuff without a tail; conservative dissection in the hilum; preservation of the left lung during anastomosis; successive anatomizing of the bronchus, the pulmonary vein, and the pulmonary artery; and one operator. Results Transplants were performed in 42 rats with a successful rate of 95.23% (40/42). The mean duration for the complete procedure was 82.93 ± 14.56 minutes. All anastomoses were completed in one attempt without vessel laceration, twisting or angulation. In our study, two animals died within three days and one animal died ten days after the operation. All grafts were well inflated with robust blood perfusion and functioned normally as demonstrated by blood gas analysis. Conclusions We have developed a modified orthotopic LT technique that can be easily performed while overcoming major drawbacks. The modified technique has many advantages, such as easy graft implanting, shortened operation time, fewer complications and high reproducibility.
Collapse
Affiliation(s)
- Haizhou Guo
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No,1277, Jiefang Avenue, Wuhan, Hubei Province, 430022, PR, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Banerjee B, Musk M, Sutanto EN, Yerkovich ST, Hopkins P, Knight DA, Lindsey-Temple S, Stick SM, Kicic A, Chambers DC. Regional differences in susceptibiity of bronchial epithelium to mesenchymal transition and inhibition by the macrolide antibiotic azithromycin. PLoS One 2012; 7:e52309. [PMID: 23284981 PMCID: PMC3528745 DOI: 10.1371/journal.pone.0052309] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 11/16/2012] [Indexed: 12/11/2022] Open
Abstract
Objective Dysregulated repair following epithelial injury is a key forerunner of disease in many organs, and the acquisition of a mesenchymal phenotype by the injured epithelial cells (epithelial to mesenchymal transition, EMT) may serve as a source of fibrosis. The macrolide antibiotic azithromycin and the DNA synthesis inhibitor mycophenolate are in clinical use but their mechanism of action remains unknown in post-transplant bronchiolitis obliterans syndrome (BOS). Here we determined if regional variation in the EMT response to TGFβ1 underlies the bronchiolocentric fibrosis leading to BOS and whether EMT could be inhibited by azithromycin or mycophenolate. Methods/Results We found that small and large airway epithelial cells from stable lung transplant patients underwent EMT when stimulated with TGFβ1, however mesenchymal protein expression was higher and loss of epithelial protein expression more complete in small airway epithelial cells. This regional difference was not mediated by changes in expression of the TGFβRII or Smad3 activation. Azithromycin potentially inhibited EMT in both small and large airway epithelial cells by inhibiting Smad3 expression, but not activation. Conclusion Collectively, these observations provide a biologic basis for a previously unexplained but widely observed clinical phenomena, and a platform for the development of new approaches to fibrotic diseases.
Collapse
Affiliation(s)
- Balarka Banerjee
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, the University of Western Australia, Nedlands, Western Australia, Australia
- Western Australia Lung Transplant Program, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Michael Musk
- Western Australia Lung Transplant Program, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Erika N. Sutanto
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | | | - Peter Hopkins
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Darryl A. Knight
- University of British Columbia, James Hogg Research Centre for Cardiovascular and Pulmonary Research, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanna Lindsey-Temple
- Centre for Asthma and Allergy Research Institute (CAARR) The Lung Institute of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Stephen M. Stick
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | - Anthony Kicic
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | - Daniel C. Chambers
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
48
|
De Paepe ME, Chu S, Hall S, Heger NE, Thanos C, Mao Q. The human fetal lung xenograft: validation as model of microvascular remodeling in the postglandular lung. Pediatr Pulmonol 2012; 47:1192-203. [PMID: 22811288 PMCID: PMC3504188 DOI: 10.1002/ppul.22617] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/23/2012] [Accepted: 04/25/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND Coordinated remodeling of epithelium and vasculature is essential for normal postglandular lung development. The value of the human-to-rodent lung xenograft as model of fetal microvascular development remains poorly defined. AIM The aim of this study was to determine the fate of the endogenous (human-derived) microvasculature in fetal lung xenografts. METHODS Lung tissues were obtained from spontaneous pregnancy losses (14-22 weeks' gestation) and implanted in the renal subcapsular or dorsal subcutaneous space of SCID-beige mice (T, B, and NK-cell-deficient) and/or nude rats (T-cell-deficient). Informed parental consent was obtained. Lung morphogenesis, microvascular angiogenesis, and epithelial differentiation were assessed at 2 and 4 weeks post-transplantation by light microscopy, immunohistochemical, and gene expression studies. Archival age-matched postmortem lungs served as control. RESULTS The vascular morphology, density, and proliferation of renal subcapsular grafts in SCID-beige mice were similar to age-matched control lungs, with preservation of the physiologic association between epithelium and vasculature. The microvasculature of subcutaneous grafts in SCID-beige mice was underdeveloped and dysmorphic, associated with significantly lower VEGF, endoglin, and angiopoietin-2 mRNA expression than renal grafts. Grafts at both sites displayed mild airspace dysplasia. Renal subcapsular grafts in nude rats showed frequent infiltration by host lymphocytes and obliterating bronchiolitis-like changes, associated with markedly decreased endogenous angiogenesis. CONCLUSION This study demonstrates the critical importance of host and site selection to ensure optimal xenograft development. When transplanted to severely immune suppressed, NK-cell-deficient hosts and engrafted in the renal subcapsular site, the human-to-rodent fetal lung xenograft provides a valid model of postglandular microvascular lung remodeling.
Collapse
Affiliation(s)
- Monique E De Paepe
- Department of Pathology, Women and Infants Hospital, Brown University, Providence, RI, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Lin X, Li W, Lai J, Okazaki M, Sugimoto S, Yamamoto S, Wang X, Gelman AE, Kreisel D, Krupnick AS. Five-year update on the mouse model of orthotopic lung transplantation: Scientific uses, tricks of the trade, and tips for success. J Thorac Dis 2012; 4:247-58. [PMID: 22754663 DOI: 10.3978/j.issn.2072-1439.2012.06.02] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 06/05/2012] [Indexed: 12/23/2022]
Abstract
It has been 5 years since our team reported the first successful model of orthotopic single lung transplantation in the mouse. There has been great demand for this technique due to the obvious experimental advantages the mouse offers over other large and small animal models of lung transplantation. These include the availability of mouse-specific reagents as well as knockout and transgenic technology. Our laboratory has utilized this mouse model to study both immunological and non-immunological mechanisms of lung transplant physiology while others have focused on models of chronic rejection. It is surprising that despite our initial publication in 2007 only few other laboratories have published data using this model. This is likely due to the technical complexity of the surgical technique and perioperative complications, which can limit recipient survival. As two of the authors (XL and WL) have a combined experience of over 2500 left and right single lung transplants, this review will summarize their experience and delineate tips and tricks necessary for successful transplantation. We will also describe technical advances made since the original description of the model.
Collapse
|
50
|
Suzuki H, Fan L, Wilkes DS. Development of obliterative bronchiolitis in a murine model of orthotopic lung transplantation. J Vis Exp 2012:3947. [PMID: 22805894 PMCID: PMC3476401 DOI: 10.3791/3947] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Orthotopic lung transplantation in rats was first reported by Asimacopoulos and colleagues in 1971 1. Currently, this method is well accepted and standardized not only for the study of allo-rejection but also between syngeneic strains for examining mechanisms of ischemia-reperfusion injury after lung transplantation. Although the application of the rat and other large animal model 2 contributed significantly to the elucidation of these studies, the scope of those investigations is limited by the scarcity of knockout and transgenic rats. Due to no effective therapies for obliterative bronchiolitis, the leading cause of death in lung transplant patients, there has been an intensive search for pre-clinical models that replicate obliterative bronchiolitis. The tracheal allograft model is the most widely used and may reproduce some of the histopathologic features of obliterative bronchiolitis 3. However, the lack of an intact vasculature with no connection to the recipient's conducting airways, and incomplete pathologic features of obliterative bronchiolitis limit the utility of this model 4. Unlike transplantation of other solid organs, vascularized mouse lung transplants have only recently been reported by Okazaki and colleagues for the first time in 2007 5. Applying the basic principles of the rat lung transplant, our lab initiated the obliterative bronchiolitis model using minor histoincompatible antigen murine orthotopic single-left lung transplants which allows the further study of obliterative bronchiolitis immunopathogenesis6.
Collapse
Affiliation(s)
- Hidemi Suzuki
- Departments of Medicine, Microbiology and Immunology, Indiana University School of Medicine, IN, USA
| | | | | |
Collapse
|