1
|
Moshkelgosha S, Levy L, Safavi S, Karunagaran S, Wilson G, Renaud-Picard B, Madu G, Ramchandani R, Oliver J, Watanabe T, Bei KF, Joe B, Li Q, Huszti E, Cheung M, Hedley D, Yeung J, Keshavjee S, Martinu T, Juvet S. Emergence of a senescent and inflammatory pulmonary CD4 + T cell population prior to lung allograft failure. SCIENCE ADVANCES 2025; 11:eadp9052. [PMID: 40117366 PMCID: PMC11927631 DOI: 10.1126/sciadv.adp9052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025]
Abstract
Survival after lung transplantation is limited by chronic lung allograft dysfunction (CLAD), an alloimmune fibrotic process leading to death or retransplantation after a median of 6 years. Immunosuppression fails to prevent CLAD, suggesting the existence of drug-resistant alloimmune pathways. We used time-of-flight mass cytometry to identify cells enriched in the bronchoalveolar lavage of patients with subsequent acute lung allograft dysfunction (ALAD), a risk factor for CLAD. We show that CD4+CD57+PD1+ T cells emerge in stable patients, conferring risks for ALAD, CLAD, and death. These cells are senescent, secrete inflammatory cytokines, and fall into two oligoclonal subsets with putative cytotoxic and follicular helper functions. Last, they are associated with fibrosis in mouse and human lung allografts, where they localize near airway epithelium and B cells. Together, our findings reveal an inflammatory T cell population that predicts future lung allograft dysfunction and may represent a rational therapeutic target.
Collapse
Affiliation(s)
- Sajad Moshkelgosha
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Liran Levy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shahideh Safavi
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Barts Health NHS Trust, London, UK
| | - Sumiha Karunagaran
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Gavin Wilson
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Benjamin Renaud-Picard
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Nouvel Hôpital Civil, Strasbourg, France
| | - Goodness Madu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Rashi Ramchandani
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Jillian Oliver
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Tatsuaki Watanabe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Ke Fan Bei
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Betty Joe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Qixuan Li
- Biostatistics Department, University Health Network, Toronto, ON, Canada
| | - Ella Huszti
- Biostatistics Department, University Health Network, Toronto, ON, Canada
| | - May Cheung
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - David Hedley
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jonathan Yeung
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Tereza Martinu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Stephen Juvet
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
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
|
3
|
Renaud-Picard B, Berra G, Hwang D, Huszti E, Miyamoto E, Berry GJ, Pal P, Juvet S, Keshavjee S, Martinu T. Spectrum of chronic lung allograft dysfunction pathology in human lung transplantation. J Heart Lung Transplant 2024; 43:1701-1715. [PMID: 38663465 DOI: 10.1016/j.healun.2024.04.002] [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: 08/23/2023] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Long-term survival after lung transplantation (LTx) remains limited by chronic lung allograft dysfunction (CLAD), which includes 2 main phenotypes: bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS), with possible overlap. We aimed to detail and quantify pathological features of these CLAD sub-types. METHODS Peripheral and central paraffin-embedded explanted lung samples were obtained from 20 consecutive patients undergoing a second LTx for CLAD, from 3 lobes. Thirteen lung samples, collected from non-transplant lobectomies or donor lungs, were used as controls. Blinded semi-quantitative grading was performed to assess airway fibrotic changes, parenchymal and pleural fibrosis, and epithelial and vascular abnormalities. RESULTS CLAD lung samples had higher scores for all airway- and lung-related parameters compared to controls. There was a notable overlap in histologic scores between BOS and RAS, with a wide range of scores in both conditions. Parenchymal and vascular fibrosis scores were significantly higher in RAS compared to BOS (p = 0.003 for both). We observed a significant positive correlation between the degree of inflammation around each airway, the severity of epithelial changes, and airway fibrosis. Immunofluorescence staining demonstrated a trend toward a lower frequency of club cells in CLAD and a higher frequency of apoptotic club cells in BOS samples (p = 0.01). CONCLUSIONS CLAD is a spectrum of airway, parenchymal, and pleural fibrosis, as well as epithelial, vascular, and inflammatory pathologic changes, where BOS and RAS overlap significantly. Our semi-quantitative grading score showed a generally high inter-reader reliability and may be useful for future CLAD histologic assessments.
Collapse
Affiliation(s)
- Benjamin Renaud-Picard
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; INSERM Unité Mixte de Recherche 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Gregory Berra
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Service de Pneumologie, Département de Médecine, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - David Hwang
- Department of Pathology, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Ei Miyamoto
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Prodipto Pal
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Kawashima M, Oliver JD, Watanabe T, Oishi H, Huang N, Konoeda C, Hirayama S, Hwang DM, Li Q, Huszti E, Liu M, Keshavjee S, Juvet S, Martinu T. Surgeon-dependent histopathological variations in minor alloantigen-mismatched mouse lung transplantation. JHLT OPEN 2024; 4:100050. [PMID: 40144241 PMCID: PMC11935354 DOI: 10.1016/j.jhlto.2023.100050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background The mouse orthotopic single lung transplant (LTx) model is an important scientific tool to explore LTx immunology. C57BL/10J (B10, H-2b) to C57BL/6J (B6, H-2b) minor alloantigen-mismatched LTx exhibits mild acute rejection and chronic fibrosis, mimicking human LTx, where acute rejection is dampened by immunosuppressants and chronic lung allograft dysfunction (CLAD) develops over time. However, we have observed variations in allograft histology across experiments, which were not explained by animal vendor or experimental conditions. The purpose of this study was to evaluate those variations objectively. Methods We performed a retrospective review of B10-to-B6 LTx performed in our laboratory 2012-2019. Only LTx without experimental interventions (eg, immunomodulatory agents or genetic modifications) examined at day 28 was eligible for this study. Mice from each surgeon were selected and divided into 3 groups to represent early, middle, and late timepoints in their mouse LTx experience (143 LTx from 5 surgeons). Histology from these LTx was graded in a randomized and blinded manner. Pathological variations and trajectories were graphed; logistic regression analyses were performed for statistical assessment. Results Distribution and trajectories of pathological outcomes were significantly different across surgeons. In multivariable logistic regression analyses, surgeon was associated with pathological outcomes whereas case number was not. Longer warm ischemia time was associated with more severe pleural fibrosis. Conclusions The B10 to B6 single LTx model can be a powerful tool to recapitulate CLAD-like histology. However, this is a challenging operation and surgeon-dependent variability in histopathological findings needs to be taken into account when designing experimental protocols.
Collapse
Affiliation(s)
- Mitsuaki Kawashima
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jillian D. Oliver
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Hisashi Oishi
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Ning Huang
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Chihiro Konoeda
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shin Hirayama
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David M. Hwang
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Qixuan Li
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Heigl T, Kaes J, Aelbrecht C, Serré J, Yamada Y, Geudens V, Van Herck A, Vanstapel A, Sacreas A, Ordies S, Frick A, Saez Gimenez B, Van Slambrouck J, Beeckmans H, Acet Oztürk NA, Orlitova M, Vaneylen A, Claes S, Schols D, Vande Velde G, Schupp J, Kaminski N, Boesch M, Korf H, van der Merwe S, Dupont L, Vanoirbeek J, Godinas L, Van Raemdonck DE, Janssens W, Gayan-Ramirez G, Ceulemans LJ, McDonough JE, Verbeken EK, Vos R, Vanaudenaerde BM. The nature of chronic rejection after lung transplantation: a murine orthotopic lung transplant study. Front Immunol 2024; 15:1369536. [PMID: 38736881 PMCID: PMC11084670 DOI: 10.3389/fimmu.2024.1369536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/25/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Chronic rejection is a major complication post-transplantation. Within lung transplantation, chronic rejection was considered as airway centred. Chronic Lung Allograft Dysfunction (CLAD), defined to cover all late chronic complications, makes it more difficult to understand chronic rejection from an immunological perspective. This study investigated the true nature, timing and location of chronic rejection as a whole, within mouse lung transplantation. Methods 40 mice underwent an orthotopic left lung transplantation, were sacrificed at day 70 and evaluated by histology and in vivo µCT. For timing and location of rejection, extra grafts were sacrificed at day 7, 35, 56 and investigated by ex vivo µCT or single cell RNA (scRNA) profiling. Results Chronic rejection originated as innate inflammation around small arteries evolving toward adaptive organization with subsequent end-arterial fibrosis and obliterans. Subsequently, venous and pleural infiltration appeared, followed by airway related bronchiolar folding and rarely bronchiolitis obliterans was observed. Ex vivo µCT and scRNA profiling validated the time, location and sequence of events with endothelial destruction and activation as primary onset. Conclusion Against the current belief, chronic rejection in lung transplantation may start as an arterial response, followed by responses in venules, pleura, and, only in the late stage, bronchioles, as may be seen in some but not all patients with CLAD.
Collapse
Affiliation(s)
- Tobias Heigl
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Celine Aelbrecht
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Jef Serré
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Yoshito Yamada
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Anke Van Herck
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
- Translational Cell and Tissue Research, KU Leuven and UZ Gasthuisberg, Leuven, Belgium
| | - Annelore Sacreas
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Sofie Ordies
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Anna Frick
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Berta Saez Gimenez
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
- Pulmonology Service, Lung Transplant Program, Hospital Universitari Vall d’Hebrón, Barcelona, Spain
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Nilüfer A. Acet Oztürk
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
- Department of Respiratory Medicine, Uludag University Faculty of Medicine, Bursa, Türkiye
| | - Michaela Orlitova
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Annemie Vaneylen
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Sandra Claes
- Translational Cell and Tissue Research, KU Leuven and UZ Gasthuisberg, Leuven, Belgium
| | - Dominique Schols
- Translational Cell and Tissue Research, KU Leuven and UZ Gasthuisberg, Leuven, Belgium
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI/MoSAIC, KU Leuven, Leuven, Belgium
| | - Jonas Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Markus Boesch
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Schalk van der Merwe
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Lieven Dupont
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Jeroen Vanoirbeek
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Laurent Godinas
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Dirk E. Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Wim Janssens
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - John E. McDonough
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Erik K. Verbeken
- Translational Cell and Tissue Research, KU Leuven and UZ Gasthuisberg, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery, KULeuven and UZ Gasthuisberg, Leuven, Belgium
| |
Collapse
|
6
|
Pavlisko EN, Adam BA, Berry GJ, Calabrese F, Cortes-Santiago N, Glass CH, Goddard M, Greenland JR, Kreisel D, Levine DJ, Martinu T, Verleden SE, Weigt SS, Roux A. The 2022 Banff Meeting Lung Report. Am J Transplant 2024; 24:542-548. [PMID: 37931751 DOI: 10.1016/j.ajt.2023.10.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
The Lung Session of the 2022 16th Banff Foundation for Allograft Pathology Conference-held in Banff, Alberta-focused on non-rejection lung allograft pathology and novel technologies for the detection of allograft injury. A multidisciplinary panel reviewed the state-of-the-art of current histopathologic entities, serologic studies, and molecular practices, as well as novel applications of digital pathology with artificial intelligence, gene expression analysis, and quantitative image analysis of chest computerized tomography. Current states of need as well as prospective integration of the aforementioned tools and technologies for complete assessment of allograft injury and its impact on lung transplant outcomes were discussed. Key conclusions from the discussion were: (1) recognition of limitations in current standard of care assessment of lung allograft dysfunction; (2) agreement on the need for a consensus regarding the standardized approach to the collection and assessment of pathologic data, inclusive of all lesions associated with graft outcome (eg, non-rejection pathology); and (3) optimism regarding promising novel diagnostic modalities, especially minimally invasive, which should be integrated into large, prospective multicenter studies to further evaluate their utility in clinical practice for directing personalized therapies to improve graft outcomes.
Collapse
Affiliation(s)
- Elizabeth N Pavlisko
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA.
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Gerald J Berry
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Nahir Cortes-Santiago
- Department of Pathology and Immunology, Texas Children's Hospital, Houston, Texas, USA
| | - Carolyn H Glass
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Martin Goddard
- Pathology Department, Royal Papworth Hospital, NHS Trust, Papworth Everard, Cambridge, UK
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, USA; Veterans Affairs Health Care System, San Francisco, California, USA
| | - Daniel Kreisel
- Department of Surgery, Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Deborah J Levine
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, California, USA
| | - Tereza Martinu
- Division of Respirology, Department of Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada; Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Stijn E Verleden
- Lung Transplant Unit, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of ASTARC, University of Antwerp, Wilrijk, Belgium
| | - S Sam Weigt
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Antoine Roux
- Department of Respiratory Medicine, Foch Hospital, Suresnes, France
| |
Collapse
|
7
|
Cui Y, Lv Z, Yang Z, Lei J. Inhibition of Prostaglandin-Degrading Enzyme 15-PGDH Mitigates Acute Murine Lung Allograft Rejection. Lung 2023; 201:591-601. [PMID: 37934242 DOI: 10.1007/s00408-023-00651-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Acute rejection is a frequent complication among lung transplant recipients and poses substantial therapeutic challenges. 15-hydroxyprostaglandin dehydrogenase (15-PGDH), an enzyme responsible for the inactivation of prostaglandin E2 (PGE2), has recently been implicated in inflammatory lung diseases. However, the role of 15-PGDH in lung transplantation rejection remains elusive. The present study was undertaken to examine the expression of 15-PGDH in rejected lung allografts and whether inhibition of 15-PGDH ameliorates acute lung allograft rejection. METHODS Orthotopic mouse lung transplantations were performed between donor and recipient mice of the same strain or allogeneic mismatched pairs. The expression of 15-PGDH in mouse lung grafts was measured. The efficacy of a selective 15-PGDH inhibitor (SW033291) in ameliorating acute rejection was assessed through histopathological examination, micro-CT imaging, and pulmonary function tests. Additionally, the mechanism underlying the effects of SW033291 treatment was explored using CD8+ T cells isolated from mouse lung allografts. RESULTS Increased 15-PGDH expression was observed in rejected allografts and allogeneic CD8+ T cells. Treatment with SW033291 led to an accumulation of PGE2, modulation of CD8+ T-cell responses and mitochondrial activity, and improved allograft function and survival. CONCLUSION Our study provides new insights into the role of 15-PGDH in acute lung rejection and highlights the therapeutic potential of inhibiting 15-PGDH for enhancing graft survival. The accumulation of PGE2 and modulation of CD8+ T-cell responses represent potential mechanisms underlying the benefits of 15-PGDH inhibition in this model. Our findings provide impetus for further exploring 15-PGDH as a target for improving lung transplantation outcomes.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, #10 Xi Tou Tiao, You An Men Wai, Fengtai, Beijing, 100069, People's Republic of China.
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, #10 Xi Tou Tiao, You An Men Wai, Fengtai, Beijing, 100069, People's Republic of China
| | - Zeran Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Jianfeng Lei
- Research Core Facilities, Capital Medical University, Beijing, 100069, People's Republic of China
| |
Collapse
|
8
|
Yombo DJK, Madala SK, Vemulapalli CP, Ediga HH, Hardie WD. Pulmonary fibroelastosis - A review. Matrix Biol 2023; 124:1-7. [PMID: 37922998 PMCID: PMC10841596 DOI: 10.1016/j.matbio.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Elastin is a long-lived fibrous protein that is abundant in the extracellular matrix of the lung. Elastic fibers provide the lung the characteristic elasticity during inhalation with recoil during exhalation thereby ensuring efficient gas exchange. Excessive deposition of elastin and other extracellular matrix proteins reduces lung compliance by impairing ventilation and compromising gas exchange. Notably, the degree of elastosis is associated with the progressive decline in lung function and survival in patients with interstitial lung diseases. Currently there are no proven therapies which effectively reduce the elastin burden in the lung nor prevent dysregulated elastosis. This review describes elastin's role in the healthy lung, summarizes elastosis in pulmonary diseases, and evaluates the current understanding of elastin regulation and dysregulation with the goal of guiding future research efforts to develop novel and effective therapies.
Collapse
Affiliation(s)
- Dan J K Yombo
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Satish K Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio USA
| | - Chanukya P Vemulapalli
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio USA
| | - Harshavardhana H Ediga
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio USA
| | - William D Hardie
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, OH, USA.
| |
Collapse
|
9
|
Watanabe T, Juvet SC, Berra G, Havlin J, Zhong W, Boonstra K, Daigneault T, Horie M, Konoeda C, Teskey G, Guan Z, Hwang DM, Liu M, Keshavjee S, Martinu T. Donor IL-17 receptor A regulates LPS-potentiated acute and chronic murine lung allograft rejection. JCI Insight 2023; 8:e158002. [PMID: 37937643 PMCID: PMC10721268 DOI: 10.1172/jci.insight.158002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 09/15/2023] [Indexed: 11/09/2023] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is a major complication after lung transplantation that results from a complex interplay of innate inflammatory and alloimmune factors, culminating in parenchymal and/or obliterative airway fibrosis. Excessive IL-17A signaling and chronic inflammation have been recognized as key factors in these pathological processes. Herein, we developed a model of repeated airway inflammation in mouse minor alloantigen-mismatched single-lung transplantation. Repeated intratracheal LPS instillations augmented pulmonary IL-17A expression. LPS also increased acute rejection, airway epithelial damage, and obliterative airway fibrosis, similar to human explanted lung allografts with antecedent episodes of airway infection. We then investigated the role of donor and recipient IL-17 receptor A (IL-17RA) in this context. Donor IL-17RA deficiency significantly attenuated acute rejection and CLAD features, whereas recipient IL-17RA deficiency only slightly reduced airway obliteration in LPS allografts. IL-17RA immunofluorescence positive staining was greater in human CLAD lungs compared with control human lung specimens, with localization to fibroblasts and myofibroblasts, which was also seen in mouse LPS allografts. Taken together, repeated airway inflammation after lung transplantation caused local airway epithelial damage, with persistent elevation of IL-17A and IL-17RA expression and particular involvement of IL-17RA on donor structural cells in development of fibrosis.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Stephen C. Juvet
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory Berra
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Jan Havlin
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wenshan Zhong
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Kristen Boonstra
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Tina Daigneault
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | | | - Chihiro Konoeda
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Grace Teskey
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Zehong Guan
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - David M. Hwang
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
House EL, Kim SY, Chalupa D, Hernady E, Groves AM, Johnston CJ, McGraw MD. IL-17A neutralization fails to attenuate airway remodeling and potentiates a proinflammatory lung microenvironment in diacetyl-exposed rats. Am J Physiol Lung Cell Mol Physiol 2023; 325:L434-L446. [PMID: 37642674 PMCID: PMC10639012 DOI: 10.1152/ajplung.00082.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Bronchiolitis obliterans (BO) is a devastating lung disease that can develop following inhalation exposure to certain chemicals. Diacetyl (DA) is one chemical commonly associated with BO development when inhaled at occupational levels. Previous studies in rats have shown that repetitive DA vapor exposures increased lung CD4+CD25+ T cells and bronchoalveolar (BAL) interleukin-17A (IL-17A) concentrations concurrent with the development of airway remodeling. We hypothesized that IL-17A neutralization would attenuate the severity of airway remodeling after repetitive DA vapor exposures. Sprague-Dawley rats were exposed to 200 parts-per-million DA vapor or filtered air (RA) for 6 h/day × 5 days and monitored for 2 wk postexposure. Treatment with IL-17A neutralization (αIL-17A) or IgG (control) began immediately following exposures and continued twice weekly until study's end. Lungs were harvested for histology, flow cytometry, and BAL analyses. Survival, oxygen saturations, and percent weight change decreased significantly in DA-exposed versus RA-exposed rats, but did not differ significantly between DA + αIL-17A versus DA + IgG. Similarly, the number nor severity of airway lesions did not differ significantly between DA + αIL-17A versus DA + IgG rats despite the percentage of lung regulatory T cells increasing with decreased BAL IL-17A concentrations. Ashcroft scoring of the distal lung parenchyma suggested worse parenchymal remodeling in DA + αIL-17A versus DA + IgG rats with increased expression of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and nuclear factor-kappa B (NF-κB). Collectively, IL-17A neutralization in DA-exposed rats failed to attenuate airway remodeling with increased expression of pro-inflammatory cytokines TNF-α, IL-1β, and NF-κB.NEW & NOTEWORTHY Interleukin-17A (IL-17A) neutralization has shown benefit previously in preclinical models of transplant-associated bronchiolitis obliterans (BO), yet it remains unknown whether IL-17A neutralization has similar benefit for other forms of BO. Here, IL-17A neutralization fails to prevent severe airway remodeling in rats exposed repetitively to the flavoring chemical diacetyl, and instead, promotes a proinflammatory microenvironment with increased expression of TNF-α, IL-1β, and NF-κB within the lung.
Collapse
Affiliation(s)
- Emma L House
- Department of Pathology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
| | - So-Young Kim
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Eric Hernady
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
| | - Angela M Groves
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, New York, United States
| | - Carl J Johnston
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew D McGraw
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
11
|
Imani J, Liu K, Cui Y, Assaker JP, Han J, Ghosh AJ, Ng J, Shrestha S, Lamattina AM, Louis PH, Hentschel A, Esposito AJ, Rosas IO, Liu X, Perrella MA, Azzi J, Visner G, El-Chemaly S. Blocking hyaluronan synthesis alleviates acute lung allograft rejection. JCI Insight 2021; 6:142217. [PMID: 34665782 PMCID: PMC8663774 DOI: 10.1172/jci.insight.142217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Lung allograft rejection results in the accumulation of low–molecular weight hyaluronic acid (LMW-HA), which further propagates inflammation and tissue injury. We have previously shown that therapeutic lymphangiogenesis in a murine model of lung allograft rejection reduced tissue LMW-HA and was associated with improved transplant outcomes. Herein, we investigated the use of 4-Methylumbelliferone (4MU), a known inhibitor of HA synthesis, to alleviate acute allograft rejection in a murine model of lung transplantation. We found that treating mice with 4MU from days 20 to 30 after transplant was sufficient to significantly improve outcomes, characterized by a reduction in T cell–mediated lung inflammation and LMW-HA content and in improved pathology scores. In vitro, 4MU directly attenuated activation, proliferation, and differentiation of naive CD4+ T cells into Th1 cells. As 4MU has already been demonstrated to be safe for human use, we believe examining 4MU for the treatment of acute lung allograft rejection may be of clinical significance.
Collapse
Affiliation(s)
- Jewel Imani
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaifeng Liu
- Division of Pulmonary and Critical Care Medicine, Boston Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ye Cui
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Junwen Han
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Auyon J Ghosh
- Division of Pulmonary, Critical Care, and Sleep Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Julie Ng
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shikshya Shrestha
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pierce H Louis
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne Hentschel
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony J Esposito
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jamil Azzi
- Transplantation Research Center, Renal Division, and
| | - Gary Visner
- Division of Pulmonary and Critical Care Medicine, Boston Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
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
|
13
|
Pezzuto F, Lunardi F, Vadori M, Zampieri D, Casiraghi F, Azzollini N, Vuljan SE, Mammana M, Vedovelli L, Schiavon M, Gregori D, Cozzi E, Rea F, Calabrese F. Chronic lung allograft pathology lesions in two rat strain combinations. J Thorac Dis 2021; 13:2833-2843. [PMID: 34164175 PMCID: PMC8182524 DOI: 10.21037/jtd-20-3415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Chronic lung allograft dysfunction remains an obstacle to long-term survival after lung transplantation. Two phenotypes have been described: obliterative bronchiolitis and restrictive allograft syndrome. Preclinical models are essential to analyze chronic lung allograft dysfunction pathophysiology. Methods Orthotopic lung transplants from 38 Lewis into Fischer 344 (Lew→F344) and 67 Brown-Norway into Lewis (BN→Lew) rats were performed in our center in the last decade. We carefully reviewed and quantified all grafts with chronic rejection (40 cases) (18 Lew→F344, 22 BN→Lew) with the aim to investigate if histological changes of chronic lung allograft dysfunction could be also detected in rat grafts. Results All animals showed human reminiscent histological lesions. Early chronic rejection lesions were detected in BN→Lew. End-stage chronic rejection with features of obliterative bronchiolitis was observed in 33% of Lew→F344; end-stage with restrictive allograft syndrome chronic rejection in 67% and 80% of Lew→F344 and BN→Lew, respectively. BN→Lew showed higher grades of endotheliitis, vascular fibrosis, and lower grades of lymphoid aggregates than Lew→F344 (P=0.007, P=0.043, P=0.004, respectively). Conclusions Chronic rejection lesions in rat lung allografts mimic those in humans. The frequent occurrence of restrictive allograft syndrome-like lesions in BN→Lew may be related to a higher degree of mismatch in this strain combination. These animal models could allow future mechanistic studies to better understand chronic lung allograft dysfunction pathogenesis.
Collapse
Affiliation(s)
- Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Davide Zampieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Nadia Azzollini
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Mammana
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Emanuele Cozzi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| |
Collapse
|
14
|
Naikawadi RP, Green G, Jones KD, Achtar-Zadeh N, Mieleszko JE, Arnould I, Kukreja J, Greenland JR, Wolters PJ. Airway Epithelial Telomere Dysfunction Drives Remodeling Similar to Chronic Lung Allograft Dysfunction. Am J Respir Cell Mol Biol 2020; 63:490-501. [PMID: 32551854 DOI: 10.1165/rcmb.2019-0374oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Telomere dysfunction is associated with multiple fibrotic lung processes, including chronic lung allograft dysfunction (CLAD)-the major limitation to long-term survival following lung transplantation. Although shorter donor telomere lengths are associated with an increased risk of CLAD, it is unknown whether short telomeres are a cause or consequence of CLAD pathology. Our objective was to test whether telomere dysfunction contributes to the pathologic changes observed in CLAD. Histopathologic and molecular analysis of human CLAD lungs demonstrated shortened telomeres in lung epithelial cells quantified by teloFISH, increased numbers of surfactant protein C immunoreactive type II alveolar epithelial cells, and increased expression of senescence markers (β-galactosidase, p16, p53, and p21) in lung epithelial cells. TRF1F/F (telomere repeat binding factor 1 flox/flox) mice were crossed with tamoxifen-inducible SCGB1a1-cre mice to generate SCGB1a1-creTRF1F/F mice. Following 9 months of tamoxifen-induced deletion of TRF1 in club cells, mice developed mixed obstructive and restrictive lung physiology, small airway obliteration on microcomputed tomography, a fourfold decrease in telomere length in airway epithelial cells, collagen deposition around bronchioles and adjacent lung parenchyma, increased type II aveolar epithelial cell numbers, expression of senescence-associated β-galactosidase in epithelial cells, and decreased SCGB1a1 expression in airway epithelial cells. These findings demonstrate that telomere dysfunction isolated to airway epithelial cells leads to airway-centric lung remodeling and fibrosis similar to that observed in patients with CLAD and suggest that lung epithelial cell telomere dysfunction may be a molecular driver of CLAD.
Collapse
Affiliation(s)
- Ram P Naikawadi
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Gary Green
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | | | - Natalia Achtar-Zadeh
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Julia E Mieleszko
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Isabel Arnould
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California; and
| | - John R Greenland
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine.,Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| |
Collapse
|
15
|
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: 3.4] [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
|
16
|
Renaud-Picard B, Vallière K, Toussaint J, Kreutter G, El-Habhab A, Kassem M, El-Ghazouani F, Olland A, Hirschi S, Porzio M, Chenard MP, Toti F, Kessler L, Kessler R. Epithelial-mesenchymal transition and membrane microparticles: Potential implications for bronchiolitis obliterans syndrome after lung transplantation. Transpl Immunol 2020; 59:101273. [PMID: 32097721 DOI: 10.1016/j.trim.2020.101273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Long term survival post lung transplantation (LTx) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). One mechanism involved is the epithelial-mesenchymal transition (EMT). Membrane microparticles (MPs) are known to be involved in some respiratory diseases and in other organs allograft rejection episodes. We hypothesized that leukocyte-derived MPs likely contribute to EMT. To emphasize this physiological concept, our objectives were to: (1) confirm the presence of EMT on explanted lungs from patients who underwent a second LTx for BOS; 2) characterize circulating MPs in transplanted patients, with or without BOS; (3) evaluate in vitro the effect of monocyte-derived MPs in EMT of human bronchial epithelial cells. Our IHC analysis on explanted graft lungs revealed significant pathological signs of EMT with an inhomogeneous destruction of the bronchial epithelium, with decreased expression of the epithelial protein E-cadherin and increased expression of the mesenchymal protein Vimentin. The immunophenotyping of MPs demonstrated that the concentration of MPs carrying E-cadherin was lower in patients affected by BOS (p = .007). In vitro, monocyte-derived MPs produced with LPS were associated with decreased E-cadherin expression (p < .05) along with significant morphological and functional cell modifications. MPs may play a role in EMT onset in bronchial epithelium following LTx.
Collapse
Affiliation(s)
- Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France.
| | - Kevin Vallière
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Justine Toussaint
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Guillaume Kreutter
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Ali El-Habhab
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Mohamad Kassem
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Fatiha El-Ghazouani
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France; Department of Thoracic Surgery and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Sandrine Hirschi
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Michele Porzio
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | | | - Florence Toti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Laurence Kessler
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| | - Romain Kessler
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| |
Collapse
|
17
|
Watanabe T, Martinu T, Chruscinski A, Boonstra K, Joe B, Horie M, Guan Z, Bei KF, Hwang DM, Liu M, Keshavjee S, Juvet SC. A B cell-dependent pathway drives chronic lung allograft rejection after ischemia-reperfusion injury in mice. Am J Transplant 2019; 19:3377-3389. [PMID: 31365766 DOI: 10.1111/ajt.15550] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) limits long-term survival after lung transplant (LT). Ischemia-reperfusion injury (IRI) promotes chronic rejection (CR) and CLAD, but the underlying mechanisms are not well understood. To examine mechanisms linking IRI to CR, a mouse orthotopic LT model using a minor alloantigen strain mismatch (C57BL/10 [B10, H-2b ] → C57BL/6 [B6, H-2b ]) and isograft controls (B6→B6) was used with antecedent minimal or prolonged graft storage. The latter resulted in IRI with subsequent airway and parenchymal fibrosis in prolonged storage allografts but not isografts. This pattern of CR after IRI was associated with the formation of B cell-rich tertiary lymphoid organs within the grafts and circulating autoantibodies. These processes were attenuated by B cell depletion, despite preservation of allograft T cell content. Our observations suggest that IRI may promote B cell recruitment that drives CR after LT. These observations have implications for the mechanisms leading to CLAD after LT.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Andrzej Chruscinski
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kristen Boonstra
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Betty Joe
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Miho Horie
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Zehong Guan
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ke Fan Bei
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David M Hwang
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Stephen C Juvet
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Guo Y, Wang Q, Li D, Onyema OO, Mei Z, Manafi A, Banerjee A, Mahgoub B, Stoler MH, Barker TH, Wilkes DS, Gelman AE, Kreisel D, Krupnick AS. Vendor-specific microbiome controls both acute and chronic murine lung allograft rejection by altering CD4 + Foxp3 + regulatory T cell levels. Am J Transplant 2019; 19:2705-2718. [PMID: 31278849 PMCID: PMC7919421 DOI: 10.1111/ajt.15523] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/09/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
Despite standardized postoperative care, some lung transplant patients suffer multiple episodes of acute and chronic rejection while others avoid graft problems for reasons that are poorly understood. Using an established model of C57BL/10 to C57BL/6 minor antigen mismatched single lung transplantation, we now demonstrate that the recipient microbiota contributes to variability in the alloimmune response. Specifically, mice from the Envigo facility in Frederick, Maryland contain nearly double the number of CD4+ Foxp3+ regulatory T cells (Tregs ) than mice from the Jackson facility in Bar Harbor, Maine or the Envigo facility in Indianapolis, Indiana (18 vs 9 vs 7%). Lung graft recipients from the Maryland facility thus do not develop acute or chronic rejection. Treatment with broad-spectrum antibiotics decreases Tregs and increases both acute and chronic graft rejection in otherwise tolerant strains of mice. Constitutive depletion of regulatory T cells, using Foxp3-driven expression of diphtheria toxin receptor, leads to the development of chronic rejection and supports the role of Tregs in both acute and chronic alloimmunity. Taken together, our data demonstrate that the microbiota of certain individuals may contribute to tolerance through Treg -dependent mechanisms and challenges the practice of indiscriminate broad-spectrum antibiotic use in the perioperative period.
Collapse
Affiliation(s)
- Yizhan Guo
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Qing Wang
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Dongge Li
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Oscar Okwudiri Onyema
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Zhongcheng Mei
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Amir Manafi
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Anirban Banerjee
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| | - Bayan Mahgoub
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States
| | - Mark H. Stoler
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States
| | - Thomas H. Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia United States
| | - David S. Wilkes
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Andrew E. Gelman
- Department of Surgery Washington University in St. Louis, Missouri, United States,Department of Pathology & Immunology, Washington University in St. Louis, Missouri, United States
| | - Daniel Kreisel
- Department of Surgery Washington University in St. Louis, Missouri, United States,Department of Pathology & Immunology, Washington University in St. Louis, Missouri, United States
| | - Alexander Sasha Krupnick
- Department of Surgery University of Virginia, Charlottesville, Virginia, United States,Carter Immunology Center University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|