1
|
Rao D, Huang D, Peng Z, Xiao D, Xie C, Zhu S, He H, Tang Z, Wu Z, Zhang Z. Triple role of exosomes in lung transplantation. Front Immunol 2025; 16:1544960. [PMID: 40292289 PMCID: PMC12021846 DOI: 10.3389/fimmu.2025.1544960] [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: 12/13/2024] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Exosomes are tiny vesicles secreted by the vast majority of cells and play an important role in physiological as well as pathological processes in the body. Circulating exosomes in Lung Transplant Recipients (LTxR) undergoing rejection contain mismatched Human Leukocyte Antigens (HLA) and lung-associated autoantigens (e.g., K-alpha1 microtubule protein and collagen V), which may induce autoantibodies, and the circulating exosomes trigger an immune response that results in rejection of the lung transplant recipient. This article discusses the role of exosomes in lung transplantation from three perspectives: exosomes as a biomarker for rejection after lung transplantation; the mechanism of exosome-mediated activation of the immune response; and the potential of exosomes as a therapeutic strategy.
Collapse
Affiliation(s)
- Dingyu Rao
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zongbo Peng
- The First Clinical College, Gannan Medical University, Ganzhou, China
| | - Dewang Xiao
- The First Clinical College, Gannan Medical University, Ganzhou, China
| | - Chunfa Xie
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shenyu Zhu
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Haoquan He
- Department of General Practice, Jinhua Jindong District Xiaoshun Town Ditian Community Health Centre, Jinhua, China
| | - Zhixian Tang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zuxiong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
2
|
Beeckmans H, Kerckhof P, Acet Öztürk N, Zajacova A, Van Slambrouck J, Bos S, Vermant M, Van Dieren LO, Goeminne T, Vandervelde C, Bardyn J, Willems E, Lauriers S, Brusselmans M, Langenhoven LV, Emonds MP, De Pelsmaeker S, Kerkhofs J, Sadeleer LD, Godinas L, Dupont LJ, Raemdonck DEV, Ceulemans LJ, Vanaudenaerde BM, Vos R. Clinical predictors for restrictive allograft syndrome: a nested case-control study. Am J Transplant 2025:S1600-6135(25)00042-5. [PMID: 39892791 DOI: 10.1016/j.ajt.2025.01.036] [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: 07/24/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Risk factors for restrictive allograft syndrome (RAS), a severe phenotype of chronic lung allograft dysfunction (CLAD) after lung transplantation, are currently not well known. In this retrospective nested case-control-study, we analyzed 69 patients with RAS and 69 matched non-CLAD controls to identify clinical risk factors for RAS. Patients with RAS demonstrated overall higher blood eosinophils (P = .02), increased bronchoalveolar eosinophils (P < .001) and lymphocytes (P = .03), and higher incidence of infections, particularly Pseudomonas species infection (P = .003), invasive fungal disease (P < .001, mainly due to Aspergillus species), SARS-CoV-2 (P < .001), and cytomegalovirus infection (P = .04), compared with non-CLAD controls. Antihuman leukocyte antigen (anti-HLA) antibodies, especially persistent donor-specific antibodies (P < 0.001), specifically targeting HLA-DQ and HLA-DR loci, and antibody-mediated rejection (P < .001), were strongly associated with later RAS. Histopathologic lung injury patterns on transbronchial biopsy (P < .001), and persistent chest computed tomography opacities in absence of pulmonary dysfunction (P < .001) were identified as early indicators of later RAS. Proactive detection and management of these risk factors could help mitigate future decline in allograft function and reduce progression to clinical RAS. Future studies should explore early treatment strategies targeting these modifiable factors to preserve allograft function and improve long-term outcomes for lung transplant recipients.
Collapse
Affiliation(s)
- Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| | - Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Nilufer Acet Öztürk
- Department of Pulmonology, Uludağ University Faculty of Medicine, Bursa, Turkey
| | - Andrea Zajacova
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Saskia Bos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Lyne O Van Dieren
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Tessa Goeminne
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Christelle Vandervelde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Josephine Bardyn
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Elisabeth Willems
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Sam Lauriers
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Marie Paule Emonds
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium; Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Steffi De Pelsmaeker
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium
| | - Johan Kerkhofs
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium
| | - Laurens De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Laurent Godinas
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Lieven J Dupont
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Kulkarni HS, Tague LK, Calabrese DR, Liao F, Liu Z, Garnica L, Shankar N, Wu X, Kulkarni DH, Bernardt C, Byers D, Chen C, Huang HJ, Witt CA, Hachem RR, Kreisel D, Atkinson JP, Greenland JR, Gelman AE. Impaired complement regulation drives chronic lung allograft dysfunction after lung transplantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.17.623951. [PMID: 39605452 PMCID: PMC11601477 DOI: 10.1101/2024.11.17.623951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
A greater understanding of chronic lung allograft dysfunction (CLAD) pathobiology, the primary cause of mortality after lung transplantation, is needed to improve outcomes. The complement system links innate to adaptive immune responses and is activated early post-lung transplantation to form the C3 convertase, a critical enzyme that cleaves the central complement component C3. We hypothesized that LTx recipients with a genetic predisposition to enhanced complement activation have worse CLAD-free survival mediated through increased adaptive alloimmunity. We interrogated a known functional C3 polymorphism (C3R102G) that increases complement activation through impaired C3 convertase inactivation in two independent LTx recipient cohorts. C3R102G, identified in at least one out of three LTx recipients, was associated with worse CLAD-free survival, particularly in the subset of recipients who developed donor-specific antibodies (DSA). In a mouse orthotopic lung transplantation model, impaired recipient complement regulation resulted in more severe obstructive airway lesions when compared to wildtype controls, despite only moderate differences in graft-infiltrating effector T cells. Impaired complement regulation promoted the intragraft accumulation of memory B cells and antibody-secreting cells, resulting in increased DSA levels. In summary, genetic predisposition to complement activation is associated with B cell activation and worse CLAD-free survival.
Collapse
Affiliation(s)
- Hrishikesh S. Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Laneshia K. Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel R. Calabrese
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Fuyi Liao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhiyi Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lorena Garnica
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nishanth Shankar
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaobo Wu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Devesha H. Kulkarni
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Cory Bernardt
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Derek Byers
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine Chen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Howard J. Huang
- Department of Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Chad A. Witt
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ramsey R. Hachem
- Department of Internal Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - John P. Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John R. Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
4
|
Jaing TH, Wang YL, Chiu CC. Time to Rethink Bronchiolitis Obliterans Syndrome Following Lung or Hematopoietic Cell Transplantation in Pediatric Patients. Cancers (Basel) 2024; 16:3715. [PMID: 39518153 PMCID: PMC11545638 DOI: 10.3390/cancers16213715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Similar in histological characteristics and clinical manifestations, bronchiolitis obliterans syndrome (BOS) can develop following lung transplantation (LTx) or hematopoietic cell transplantation (HCT). In contrast to lung transplantation, where BOS is restricted to the lung allograft, HCT-related systemic graft-versus-host disease (GVHD) is the root cause of BOS. Because lung function declines following HCT, diagnosis becomes more difficult. Given the lack of proven effective medicines, treatment is based on empirical evidence. Methods: Cross-disciplinary learning is crucial, and novel therapies are under investigation to improve survival and avoid LTx. Recent advances have focused on updating the understanding of the etiology, clinical features, and pathobiology of BOS. It emphasizes the significance of learning from experts in other transplant modalities, promoting cross-disciplinary knowledge. Results: Our treatment algorithms are derived from extensive research and expert clinical input. It is important to ensure that immunosuppression is optimized and that any other conditions or contributing factors are addressed, if possible. Clear treatment algorithms are provided for each condition, drawing from the published literature and consensus clinical opinion. There are several novel therapies currently being investigated, such as aerosolized liposomal cyclosporine, Janus kinase inhibitors, antifibrotic therapies, and B-cell-directed therapies. Conclusions: We urgently need innovative treatments that can greatly increase survival rates and eliminate the need for LTx or re-transplantation.
Collapse
Affiliation(s)
- Tang-Her Jaing
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Lun Wang
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Chia-Chi Chiu
- Division of Nursing, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| |
Collapse
|
5
|
Tanaka S, Takahashi M, Kayawake H, Yutaka Y, Ohsumi A, Nakajima D, Ikezoe K, Tanizawa K, Handa T, Date H. Differences in chronic lung allograft dysfunction between deceased-donor lung transplantation and living-donor lobar lung transplantation. J Thorac Cardiovasc Surg 2024:S0022-5223(24)00913-9. [PMID: 39393626 DOI: 10.1016/j.jtcvs.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE To explore the characteristics and prognostic impact of chronic lung allograft dysfunction (CLAD) after deceased-donor lung transplantation and living-donor lobar lung transplantation, wherein the lower lobes from 2 donors are usually transplanted into one recipient. METHODS The clinical data of 123 deceased-donor and 67 living-donor lung transplantations performed in adult patients at our institution between June 2008 and September 2019 were retrospectively reviewed. The cumulative incidence of CLAD was evaluated on a per-recipient and per-donor graft basis using the Kaplan-Meier method. RESULTS A smaller number of human leukocyte antigen mismatches, shorter ischemic time, and lower incidence of grade 3 primary graft dysfunction were observed in living-donor transplantation than in deceased-donor transplantation (P < .001). Restrictive allograft syndrome-type CLAD occurred in 9 (20.9%) of 43 patients with CLAD after deceased-donor transplantation and 9 (45.0%) of 20 patients with CLAD after living-donor transplantation. CLAD occurred unilaterally in 15 patients (75.0%) after bilateral living-donor transplantation. Despite the greater incidence of restrictive allograft syndrome-type CLAD after living-donor transplantation, the overall survival rates after the transplantation and survival rates after the onset of CLAD were comparable between the patients receiving deceased-donor transplants and living-donor transplants. The cumulative incidence of CLAD per recipient was similar between recipients of deceased-donor and the living-donor transplants (P = .32). In the per-donor graft analysis, the cumulative incidence of CLAD was significantly lower in the living-donor grafts than in the deceased-donor grafts (P = .003). CONCLUSIONS The manifestation of CLAD after living-donor lobar lung transplantation is unique and differs from that after deceased-donor lung transplantation.
Collapse
Affiliation(s)
- Satona Tanaka
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan.
| | - Mamoru Takahashi
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Hidenao Kayawake
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Yojiro Yutaka
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Kohei Ikezoe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Handa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
6
|
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
|
7
|
Milesi J, Gras D, Chanez P, Coiffard B. Airway epithelium in lung transplantation: a potential actor for post-transplant complications? Eur Respir Rev 2024; 33:240093. [PMID: 39603662 PMCID: PMC11600126 DOI: 10.1183/16000617.0093-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/20/2024] [Indexed: 11/29/2024] Open
Abstract
Lung transplantation, a critical intervention for end-stage lung diseases, is frequently challenged by post-transplant complications. Indeed, primary graft dysfunction, anastomotic complications, infections and acute and chronic rejections pose significant hurdles in lung transplantation. While evidence regarding the role of airway epithelium after lung transplantation is still emerging, its importance is becoming increasingly recognised. This review looks at the complex involvement of airway epithelium in various post-transplant complications, while emphasising the utility of airway epithelial culture as a research model. In summary, by elucidating the involvement of airway epithelium in each post-transplant complication and explaining these intricate processes, the review aims to guide specific future research efforts and therapeutic strategies aimed at improving lung transplant outcomes and enhancing patient care.
Collapse
Affiliation(s)
- Jules Milesi
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Delphine Gras
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pascal Chanez
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Benjamin Coiffard
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
8
|
Ponholzer F, Bogensperger C, Krendl FJ, Krapf C, Dumfarth J, Schneeberger S, Augustin F. Beyond the organ: lung microbiome shapes transplant indications and outcomes. Eur J Cardiothorac Surg 2024; 66:ezae338. [PMID: 39288305 PMCID: PMC11466426 DOI: 10.1093/ejcts/ezae338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/19/2024] Open
Abstract
The lung microbiome plays a crucial role in the development of chronic lung diseases, which may ultimately lead to the need for lung transplantation. Also, perioperative results seem to be connected with altered lung microbiomes and its dynamic changes providing a possible target for optimizing short-term outcome after transplantation. A literature review using MEDLINE, PubMed Central and Bookshelf was performed. Chronic lung allograft dysfunction (CLAD) seems to be influenced and partly triggered by changes in the pulmonary microbiome and dysbiosis, e.g. through increased bacterial load or abundance of specific species such as Pseudomonas aeruginosa. Additionally, the specific indications for transplantation, with their very heterogeneous changes and influences on the pulmonary microbiome, influence long-term outcome. Next to composition and measurable bacterial load, dynamic changes in the allografts microbiome also possess the ability to alter long-term outcomes negatively. This review discusses the "new" microbiome after transplantation and the associations with direct postoperative outcome. With the knowledge of these principles the impact of alterations in the pulmonary microbiome in hindsight to CLAD and possible therapeutic implications are described and discussed. The aim of this review is to summarize the current literature regarding pre- and postoperative lung microbiomes and how they influence different lung diseases on their progression to failure of conservative treatment. This review provides a summary of current literature for centres looking for further options in optimizing lung transplant outcomes and highlights possible areas for further research activities investigating the pulmonary microbiome in connection to transplantation.
Collapse
Affiliation(s)
- Florian Ponholzer
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Bogensperger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Julius Krendl
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Krapf
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Dumfarth
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
9
|
Sindu D, Bansal S, Buddhdev B, McAnally K, Mohamed H, Walia R, Mohanakumar T, Tokman S. Late-Onset Exudative Pleural Effusions Without Concomitant Airway Obstruction or Lung Parenchymal Abnormalities: A Novel Presentation of Chronic Lung Allograft Dysfunction. Transpl Int 2024; 37:12395. [PMID: 38357217 PMCID: PMC10866027 DOI: 10.3389/ti.2024.12395] [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: 11/08/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
Restrictive allograft syndrome (RAS) is an aggressive variant of CLAD characterized by progressive restrictive ventilatory decline and persistent pleuro-parenchymal changes that can be seen on chest CT. We identified four lung transplant recipients with a progressive restrictive ventilatory defect due to lymphocyte-predominant exudative pleural effusions, but no pleuro-parenchymal abnormalities typical of RAS. Using molecular analysis, we also found increased levels of previously described immune markers of RAS, including NFkB, 20S proteasome, lipocalin, TNFα, and TGFβ, within the circulating small extracellular vesicles of the remaining living lung transplant recipient. Despite the absence of lung parenchymal changes, these patients had a poor prognosis with rapid deterioration in allograft function and no response to pleural-based interventions such as thoracentesis, decortication, and pleurodesis. We hypothesize that these cases represent a distinct CLAD phenotype characterized by progressive restriction due to pleural inflammation, lymphocyte-predominant pleural effusion, resultant compressive atelectasis, and eventual respiratory failure in the absence of lung parenchymal involvement.
Collapse
Affiliation(s)
- Devika Sindu
- Norton Thoracic Institute, Phoenix, AZ, United States
| | | | - Bhuvin Buddhdev
- Norton Thoracic Institute, Phoenix, AZ, United States
- School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Kendra McAnally
- Norton Thoracic Institute, Phoenix, AZ, United States
- School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Hesham Mohamed
- Norton Thoracic Institute, Phoenix, AZ, United States
- School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Rajat Walia
- Norton Thoracic Institute, Phoenix, AZ, United States
- School of Medicine, Creighton University, Phoenix, AZ, United States
| | | | - Sofya Tokman
- Norton Thoracic Institute, Phoenix, AZ, United States
- School of Medicine, Creighton University, Phoenix, AZ, United States
| |
Collapse
|
10
|
Benazzo A, Bagnera C, Ius F, Del Fante C, Gottlieb J, Hoetzenecker K, Meloni F, Jaksch P, Greer M. A European Multi-Center Analysis of Extracorporeal Photopheresis as Therapy for Chronic Lung Allograft Dysfunction. Transpl Int 2024; 36:11551. [PMID: 38282747 PMCID: PMC10811603 DOI: 10.3389/ti.2023.11551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Extracorporeal photopheresis (ECP) is used by few lung transplant centers to treat chronic lung allograft dysfunction (CLAD). Although reported results suggest a beneficial effect on CLAD progression, evidence is limited to single center experiences. The aim of this study is to analyze outcomes of ECP in a large multicenter European cohort. The primary endpoint was patient survival after initiation of ECP. This study included 631 patients, 87% suffered from bronchiolitis obliterans syndrome (BOS), and 13% had restrictive allograft syndrome (RAS). Long-term stabilization was achieved in 42%, improvement in 9%, and no response in 26%. Within the first 12 months of therapy, 23% of patients died. Patients' survival after initiation of ECP at 5 years was 56% in stable, 70% in responders, and 35% in non-responders (p = 0.001). In multivariable Cox regression, both stabilization (HR: 0.48, CI: 0.27-0.86, p = 0.013) and response (HR: 0.11, CI: 0.04-0.35, p < 0.001) to ECP were associated with survival. Absolute FEV1 at baseline was also protective (HR: 0.09, CI: 0.01-0.94, p = 0.046). RAS phenotype was the only risk factor for mortality (HR: 2.11, 1.16-3.83, p = 0.006). This study provides long-term outcomes of ECP use in CLAD patients in the largest published cohort to date. Two-thirds of the cohort had a sustained response to ECP with excellent long-term results.
Collapse
Affiliation(s)
- Alberto Benazzo
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Cecilia Bagnera
- Malattie dell’Apparato Respiratorio, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fabio Ius
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Claudia Del Fante
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jens Gottlieb
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Centre for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease Hannover, Hannover, Germany
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Federica Meloni
- Malattie dell’Apparato Respiratorio, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Mark Greer
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Centre for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease Hannover, Hannover, Germany
| |
Collapse
|
11
|
Bai YZ, Kopecky BJ, Lavine KJ, Kreisel D. Ferroptosis in the post-transplantation inflammatory response. Cell Immunol 2023; 393-394:104774. [PMID: 37839157 DOI: 10.1016/j.cellimm.2023.104774] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Transplantation is a life-saving therapy for patients with end-stage organ disease. Successful outcomes after transplantation require mitigation of the post-transplant inflammatory response, limiting alloreactivity, and prevention of organ rejection. Traditional immunosuppressive regimens aim to dampen the adaptive immune response; however, recent studies have shown the feasibility and efficacy of targeting the innate immune response. Necroinflammation initiated by donor organ cell death is implicated as a critical mediator of primary graft dysfunction, acute rejection, and chronic rejection. Ferroptosis is a form of regulated cell death that triggers post-transplantation inflammation and drives the activation of both innate and adaptive immune cells. There is a growing acceptance of the clinical relevance of ferroptosis to solid organ transplantation. Modulating ferroptosis may be a potentially promising strategy to reduce complications after organ transplantation.
Collapse
Affiliation(s)
- Yun Zhu Bai
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Benjamin J Kopecky
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Kory J Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
12
|
Bansal S, Arjuna A, Franz B, Guerrero-Alba A, Canez J, Fleming T, Rahman M, Hachem R, Mohanakumar T. Extracellular vesicles: a potential new player in antibody-mediated rejection in lung allograft recipients. FRONTIERS IN TRANSPLANTATION 2023; 2:1248987. [PMID: 38993876 PMCID: PMC11235353 DOI: 10.3389/frtra.2023.1248987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/22/2023] [Indexed: 07/13/2024]
Abstract
Identification of recipients with pre-existing antibodies and cross-matching of recipient sera with donor lymphocytes have reduced the incidence of antibody-mediated rejection (AMR) after human lung transplantation. However, AMR is still common and requires not only immediate intervention but also has long-term consequences including an increased risk of chronic lung allograft dysfunction (CLAD). The mechanisms resulting in AMR remain largely unknown due to the variation in clinical and histopathological features among lung transplant recipients; however, several reports have demonstrated a strong association between the development of antibodies against mismatched donor human leucocyte antigens [donor-specific antibodies (DSAs)] and AMR. In addition, the development of antibodies against lung self-antigens (K alpha1 tubulin and collagen V) also plays a vital role in AMR pathogenesis, either alone or in combination with DSAs. In the current article, we will review the existing literature regarding the association of DSAs with AMR, along with clinical diagnostic features and current treatment options for AMR. We will also discuss the role of extracellular vesicles (EVs) in the immune-related pathogenesis of AMR, which can lead to CLAD.
Collapse
Affiliation(s)
- Sandhya Bansal
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Ashwini Arjuna
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Brian Franz
- HLA Laboratory, Vitalant, Phoenix, AZ, United States
| | - Alexa Guerrero-Alba
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Jesse Canez
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Mohammad Rahman
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Ramsey Hachem
- Department of Surgery, Washington University, St. Louis, MO, United States
| | - T. Mohanakumar
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
13
|
Tian D, Zheng X, Tang H, Huang H, Wang J, Xu L, Li C, Yan H, Yu R, Nan J, Liu M, Guo X, Jian S, Wang T, Deng S, Pu Q, Liu L. Metformin attenuates chronic lung allograft dysfunction: evidence in rat models. Respir Res 2023; 24:192. [PMID: 37516880 PMCID: PMC10386298 DOI: 10.1186/s12931-023-02492-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) directly causes an abysmal long-term prognosis after lung transplantation (LTx), but effective and safe drugs are not available. Metformin exhibits high therapeutic potential due to its antifibrotic and immunomodulatory effects; however, it is unclear whether metformin exerts a therapeutic effect in CLAD. We sought to investigate the effect of metformin on CLAD based on rat models. METHODS Allogeneic LTx rats were treated with Cyclosporin A (CsA) in the first week, followed by metformin, CsA, or vehicle treatment. Syngeneic LTx rats received only vehicles. All rats were sacrificed on post-transplant week 4. Pathology of lung graft, spleen, and thymus, extent of lung fibrosis, activity of profibrotic cytokines and signaling pathway, adaptive immunity, and AMPK activity were then studied. RESULTS Allogeneic recipients without maintenance CsA treatment manifested CLAD pathological characteristics, but these changes were not observed in rats treated with metformin. For the antifibrotic effect, metformin suppressed the fibrosis extent and profibrotic cytokine expression in lung grafts. Regarding immunomodulatory effect, metformin reduced T- and B-cell infiltration in lung grafts, spleen and thymus weights, the T- and B-cell zone areas in the spleen, and the thymic medullary area. In addition, metformin activated AMPK in lung allografts and in α-SMA+ cells and T cells in the lung grafts. CONCLUSIONS Metformin attenuates CLAD in rat models, which could be attributed to the antifibrotic and immunomodulatory effects. AMPK activation suggests the potential molecular mechanism. Our study provides an experimental rationale for further clinical trials.
Collapse
Affiliation(s)
- Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 610041, Chengdu, China
- Lung Transplant Research Laboratory, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiangyun Zheng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 610041, Chengdu, China
- Lung Transplant Research Laboratory, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Hongtao Tang
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Heng Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 610041, Chengdu, China
- Lung Transplant Research Laboratory, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Junjie Wang
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Lin Xu
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Caihan Li
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Haoji Yan
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Ruixuan Yu
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinzhu Nan
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Menggen Liu
- Heart and Lung Transplant Research Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiaoguang Guo
- Department of Pathology, Nanchong Central Hospital, Nanchong, 637000, China
| | - Shunhai Jian
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
- Department of Respiratory and Critical Care Medicine, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen, 518000, China
| | - Senyi Deng
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
14
|
Beeckmans H, Bos S, Vos R, Glanville AR. Acute Rejection and Chronic Lung Allograft Dysfunction: Obstructive and Restrictive Allograft Dysfunction. Clin Chest Med 2023; 44:137-157. [PMID: 36774160 DOI: 10.1016/j.ccm.2022.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Lung transplantation is an established treatment of well-selected patients with end-stage respiratory diseases. However, lung transplant recipients have the highest rates of acute and chronic rejection among transplanted solid organs. Owing to ongoing alloimmune recognition and associated immune-driven airway/vascular remodeling, precipitated by multifactorial, endogenous or exogenous, post-transplant injuries to the bronchovascular axis of the secondary pulmonary lobule, most lung transplant recipients will suffer from a pathophysiological decline of their allograft, either functionally and/or structurally. This review discusses current knowledge, barriers, and gaps in acute cellular rejection and chronic lung allograft dysfunction-the greatest impediment to long-term post-transplant survival.
Collapse
Affiliation(s)
- Hanne Beeckmans
- Department of Chronic Diseases and Metabolism, KU Leuven, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Leuven, Belgium
| | - Saskia Bos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium; Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, KU Leuven, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| | | |
Collapse
|
15
|
Wijbenga N, Hoek RAS, Mathot BJ, Seghers L, Moor CC, Aerts JGJV, Bos D, Manintveld OC, Hellemons ME. Diagnostic performance of electronic nose technology in chronic lung allograft dysfunction. J Heart Lung Transplant 2023; 42:236-245. [PMID: 36283951 DOI: 10.1016/j.healun.2022.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND There is a need for reliable biomarkers for the diagnosis of chronic lung allograft dysfunction (CLAD). In this light, we investigated the diagnostic value of exhaled breath analysis using an electronic nose (eNose) for CLAD, CLAD phenotype, and CLAD stage in lung transplant recipients (LTR). METHODS We performed eNose measurements in LTR with and without CLAD, visiting the outpatient clinic. Through supervised machine learning, the diagnostic value of eNose for CLAD was assessed in a random training and validation set. Next, we investigated the diagnostic value of the eNose measurements combined with known risk factors for CLAD. Model performance was evaluated using ROC-analysis. RESULTS We included 152 LTR (median age 60 years, 49% females), of whom 38 with CLAD. eNose-based classification of patients with and without CLAD provided an AUC of 0.86 in the training set, and 0.82 in the validation set. After adding established risk factors for CLAD (age, gender, type of transplantation, time after transplantation and prior occurrence of acute cellular rejection) to a model with the eNose data, the discriminative ability of the model improved to an AUC of 0.94 (p = 0.02) in the training set and 0.94 (p = 0.04) in the validation set. Discrimination between BOS and RAS was good (AUC 0.95). Discriminative ability for other phenotypes (AUCs ranging 0.50-0.92) or CLAD stages (AUC 0.56) was limited. CONCLUSION Exhaled breath analysis using eNose is a promising novel biomarker for enabling diagnosis and phenotyping CLAD. eNose technology could be a valuable addition to the diagnostic armamentarium for suspected graft failure in LTR.
Collapse
Affiliation(s)
- Nynke Wijbenga
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rogier A S Hoek
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bas J Mathot
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Leonard Seghers
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Catharina C Moor
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Olivier C Manintveld
- Department of Cardiology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Merel E Hellemons
- Department of Respiratory Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
16
|
Lynch TJ, Ahlers BA, Swatek AM, Ievlev V, Pai AC, Brooks L, Tang Y, Evans IA, Meyerholz DK, Engelhardt JF, Parekh KR. Ferret Lung Transplantation Models Differential Lymphoid Aggregate Morphology Between Restrictive and Obstructive Forms of Chronic Lung Allograft Dysfunction. Transplantation 2022; 106:1974-1989. [PMID: 35442232 PMCID: PMC9529760 DOI: 10.1097/tp.0000000000004148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Long-term survival after lung transplantation remains limited by chronic lung allograft dysfunction (CLAD). CLAD has 2 histologic phenotypes, namely obliterative bronchiolitis (OB) and restrictive alveolar fibroelastosis (AFE), which have distinct clinical presentations, pathologies, and outcomes. Understanding of OB versus AFE pathogenesis would improve with better animal models. METHODS We utilized a ferret orthotopic single-lung transplantation model to characterize allograft fibrosis as a histologic measure of CLAD. Native lobes and "No CLAD" allografts lacking aberrant histology were used as controls. We used morphometric analysis to evaluate the size and abundance of B-cell aggregates and tertiary lymphoid organs (TLOs) and their cell composition. Quantitative RNA expression of 47 target genes was performed simultaneously using a custom QuantiGene Plex Assay. RESULTS Ferret lung allografts develop the full spectrum of human CLAD histology including OB and AFE subtypes. While both OB and AFE allografts developed TLOs, TLO size and number were greater with AFE histology. More activated germinal center cells marked by B-cell lymphoma 6 Transcription Repressor, (B-cell lymphoma 6) expression and fewer cells expressing forkhead box P3 correlated with AFE, congruent with greater diffuse immunoglobulin, plasma cell abundance, and complement 4d staining. Furthermore, forkhead box P3 RNA induction was significant in OB allografts specifically. RNA expression changes were seen in native lobes of animals with AFE but not OB when compared with No CLAD native lobes. CONCLUSIONS The orthotopic ferret single-lung transplant model provides unique opportunities to better understand factors that dispose allografts to OB versus AFE. This will help develop potential immunomodulatory therapies and antifibrotic approaches for lung transplant patients.
Collapse
Affiliation(s)
- Thomas J. Lynch
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Bethany A. Ahlers
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Anthony M. Swatek
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Vitaly Ievlev
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Albert C. Pai
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Leonard Brooks
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Idil A. Evans
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Kalpaj R. Parekh
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
17
|
Santos J, Calabrese DR, Greenland JR. Lymphocytic Airway Inflammation in Lung Allografts. Front Immunol 2022; 13:908693. [PMID: 35911676 PMCID: PMC9335886 DOI: 10.3389/fimmu.2022.908693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Lung transplant remains a key therapeutic option for patients with end stage lung disease but short- and long-term survival lag other solid organ transplants. Early ischemia-reperfusion injury in the form of primary graft dysfunction (PGD) and acute cellular rejection are risk factors for chronic lung allograft dysfunction (CLAD), a syndrome of airway and parenchymal fibrosis that is the major barrier to long term survival. An increasing body of research suggests lymphocytic airway inflammation plays a significant role in these important clinical syndromes. Cytotoxic T cells are observed in airway rejection, and transcriptional analysis of airways reveal common cytotoxic gene patterns across solid organ transplant rejection. Natural killer (NK) cells have also been implicated in the early allograft damage response to PGD, acute rejection, cytomegalovirus, and CLAD. This review will examine the roles of lymphocytic airway inflammation across the lifespan of the allograft, including: 1) The contribution of innate lymphocytes to PGD and the impact of PGD on the adaptive immune response. 2) Acute cellular rejection pathologies and the limitations in identifying airway inflammation by transbronchial biopsy. 3) Potentiators of airway inflammation and heterologous immunity, such as respiratory infections, aspiration, and the airway microbiome. 4) Airway contributions to CLAD pathogenesis, including epithelial to mesenchymal transition (EMT), club cell loss, and the evolution from constrictive bronchiolitis to parenchymal fibrosis. 5) Protective mechanisms of fibrosis involving regulatory T cells. In summary, this review will examine our current understanding of the complex interplay between the transplanted airway epithelium, lymphocytic airway infiltration, and rejection pathologies.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
| | - Daniel R. Calabrese
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| | - John R. Greenland
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
18
|
Lobashevsky A, Niemann M, Kowinski B, Higgins N, Abdel-Wareth L, Atrabulsi B, Roe D, Hage C. Formation of donor-specific antibodies depends on the epitope load of mismatched hlas in lung transplant recipients: A retrospective single-center study. Clin Transplant 2022; 36:e14755. [PMID: 35771140 DOI: 10.1111/ctr.14755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/27/2022]
Abstract
The development of donor-specific antibodies (DSA) has a significant impact on graft outcome in solid organ transplantation. Mismatched HLAs are recognized directly and indirectly by the recipient immune system. Both pathways occur in parallel and result in the generation of plasma cells, DSA, cytotoxic and T helper lymphocytes. Here, we present the results of an analysis of the epitope load of mismatched HLAs in a cohort of 220 lung transplant recipients using two in silico algorithms, HLAMatchmaker and PIRCHE-II (Predicted Indirectly ReCognizable HLA Epitopes). De novo DSA (dnDSA) were detected by single antigen bead assays. The percentage of recipients who developed dnDSA was significantly higher in the group of patients who received lung transplants with a mismatching score above the detected threshold than in the group of patients who received lung transplants with a mismatching score below the threshold. In a multivariate Cox proportional hazard analysis, the PIRCHE-II score appeared to be a superior predictor of dnDSA formation. In addition, PIRCHE-II technology was shown to be useful in predicting separate dnDSA1 and dnDSA2 formation. We conclude that both algorithms can be used for the evaluation of the epitope load of mismatched HLAs and the prediction of DSA development in lung transplant recipients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andrew Lobashevsky
- Transplant Immunology Laboratory, Methodist Hospital, IUHealth Inc., Indianapolis, IN, USA.,Mubadala, National Reference Laboratory, HLA Laboratory, Abu Dhabi, UAE
| | | | - Brenda Kowinski
- Lung Transplant Program, IUHealth Inc., Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nancy Higgins
- Transplant Immunology Laboratory, Methodist Hospital, IUHealth Inc., Indianapolis, IN, USA
| | | | - Basel Atrabulsi
- Mubadala, National Reference Laboratory, HLA Laboratory, Abu Dhabi, UAE
| | - David Roe
- Lung Transplant Program, IUHealth Inc., Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chadi Hage
- McGowen Institute of Regenerative medicine, UPMC, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Sekulovski M, Simonska B, Peruhova M, Krastev B, Peshevska-Sekulovska M, Spassov L, Velikova T. Factors affecting complications development and mortality after single lung transplant. World J Transplant 2021; 11:320-334. [PMID: 34447669 PMCID: PMC8371496 DOI: 10.5500/wjt.v11.i8.320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/15/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Lung transplantation (LT) is a life-saving therapeutic procedure that prolongs survival in patients with end-stage lung disease. Furthermore, as a therapeutic option for high-risk candidates, single LT (SLT) can be feasible because the immediate morbidity and mortality after transplantation are lower compared to sequential single (double) LT (SSLTx). Still, the long-term overall survival is, in general, better for SSLTx. Despite the great success over the years, the early post-SLT period remains a perilous time for these patients. Patients who undergo SLT are predisposed to evolving early or late postoperative complications. This review emphasizes factors leading to post-SLT complications in the early and late periods including primary graft dysfunction and chronic lung allograft dysfunction, native lung complications, anastomosis complications, infections, cardiovascular, gastrointestinal, renal, and metabolite complications, and their association with morbidity and mortality in these patients. Furthermore, we discuss the incidence of malignancy after SLT and their correlation with immunosuppression therapy.
Collapse
Affiliation(s)
- Metodija Sekulovski
- Department of Anesthesiology and Intensive care, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Bilyana Simonska
- Department of Anesthesiology and Intensive care, University Hospital Lozenetz, Sofia 1407, Bulgaria
| | - Milena Peruhova
- Department of Gastroenterology, University Hospital Lozenetz, Sofia 1407, Bulgaria
| | - Boris Krastev
- Department of Clinical Oncology, MHAT Hospital for Women Health Nadezhda, Sofia 1330, Bulgaria
| | | | - Lubomir Spassov
- Department of Cardiothoracic Surgery, University Hospital Lozenetz, Sofia 1431, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia 1407, Bulgaria
| |
Collapse
|
20
|
Klouda T, Vargas SO, Midyat L. Restrictive allograft syndrome after lung transplantation. Pediatr Transplant 2021; 25:e14000. [PMID: 33728767 DOI: 10.1111/petr.14000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/07/2021] [Accepted: 02/22/2021] [Indexed: 11/29/2022]
Abstract
Despite recent advances over the past decade in lung transplantation including improved surgical technique and immunotherapy, the diagnosis and treatment of chronic lung allograft dysfunction remains a significant barrier to recipient survival. Aside from bronchiolitis obliterans syndrome, a restrictive phenotype called restrictive allograft syndrome has recently been recognized and affects up to 35% of all patients with CLAD. The main characteristics of RAS include a persistent and unexplained decline in lung function compared to baseline and persistent parenchymal infiltrates on imaging. The median survival after diagnosis of RAS is 6 to 18 months, significantly shorter than other forms of CLAD. Treatment options are limited, as therapies used for BOS are typically ineffective at halting disease progression. Specific medications such as fibrinolytics are lacking large, multicenter prospective studies. In this manuscript, we discuss the definition, mechanism, and characteristics of RAS while highlighting the similarities and differences between other forms of CLAD. We also review the diagnoses along with current and potential treatment options that are available for patients. Finally, we discuss the existing knowledge gaps and areas for future research to improve patient outcomes and understanding of RAS.
Collapse
Affiliation(s)
- Timothy Klouda
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Levent Midyat
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Eichenlaub M, Ruettner B, Seiler A, Jenewein J, Boehler A, Benden C, Wutzler U, Goetzmann L. The Actualization of the Transplantation Complex on the Axis of Psychosomatic Totality-Results of a Qualitative Study. Healthcare (Basel) 2021; 9:healthcare9040455. [PMID: 33921523 PMCID: PMC8069072 DOI: 10.3390/healthcare9040455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
Although transplantation medicine is not new, there is a clinically justified gap in the existing literature with respect to the psychological processing of lung transplants. The present study aims to examine whether lung transplantation leads to an actualization of psychological, e.g., oral-sadistic fantasies. Following a qualitative approach, 38 lung transplant patients were interviewed three times within the first six months after transplantation. Data analysis focused on identifying unconscious and conscious material. The inter-rater reliability for all codes was calculated using Krippendorff’s Alpha (c-α-binary = 0.94). Direct and implicit evidence of a so-called transplantation complex was detected e.g., regarding the “incorporation” of the dead donor and his lungs. These processes occur predominantly at an imaginary level and are related to the body. Our findings emphasize that such psychological aspects should be borne in mind in the psychological treatment of lung-transplant patients in order to improve the processing of lung transplants, and that this might have a positive effect on patient adherence.
Collapse
Affiliation(s)
- Marie Eichenlaub
- Department of Psychology, Medical School Hamburg MSH, 20457 Hamburg, Germany;
- Correspondence:
| | - Barbara Ruettner
- Department of Psychology, Medical School Hamburg MSH, 20457 Hamburg, Germany;
| | - Annina Seiler
- Department of Radiation Oncology and Competence Center for Palliative Care and Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland;
| | - Josef Jenewein
- Department of Medical Psychology and Psychotherapy, University Hospital of Graz, 8036 Graz, Austria;
| | | | - Christian Benden
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland;
| | - Uwe Wutzler
- Clinic for Psychosomatic Medicine and Psychotherapy, Asklepios Fachklinikum Stadtroda, 07646 Stadtroda, Germany;
| | - Lutz Goetzmann
- Institute of Philosophy, Psychoanalysis and Cultural Studies (IPPK), 12047 Berlin, Germany;
| |
Collapse
|
22
|
Regulatory and Effector Cell Disequilibrium in Patients with Acute Cellular Rejection and Chronic Lung Allograft Dysfunction after Lung Transplantation: Comparison of Peripheral and Alveolar Distribution. Cells 2021; 10:cells10040780. [PMID: 33916034 PMCID: PMC8065700 DOI: 10.3390/cells10040780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 01/19/2023] Open
Abstract
Background: The immune mechanisms occurring during acute rejection (AR) and chronic lung allograft dysfunction are a challenge for research and the balance between effector and regulatory cells has not been defined completely. In this study, we aimed to elucidate the interaction of effector cells, mainly Th17, Th1 and Th2, and regulatory cells including (CD4+CD25+CD127low/−) T reg cells and phenotypes of B regs, CD19+CD24hiCD38hi, CD19+CD24hiCD27hi and CD19+CD5+CD1d+. Methods: Bronchoalveolar lavage cells (BAL) and peripheral blood mononuclear cells (PBMCs) from stable lung transplanted (LTx )subjects (n = 4), AR patients (n = 6) and bronchiolitis obliterans syndrome (BOS) (n = 6) were collected at the same time. Cellular subsets were detected through flow cytometry. Results: A predominance of Th17 cells subtypes in the PBMCs and BAL and a depletion of Tregs, that resulted in decrease Treg/Th17 ratio, was observed in the AR group. CD19+CD24hiCD38hi Bregs resulted increased in BAL of AR patients. Th1 cells predominance and a reduction of Tregs cells was observed in BAL from AR patients. Moreover, multivariate analysis showed interdependences within studied variables revealing that effector cells and regulatory cells can effectively discriminate patients’ immunological status. Conclusions: In AR, BOS and stable lung transplant, regulatory and effector cells clearly demonstrated different pathways of activation. Understanding of the balance of T cells and T and B regulatory cells can offers insights into rejection.
Collapse
|
23
|
Werlein C, Ackermann M, Hoffmann TL, Laenger F, Jonigk D. [Fibrotic remodeling of the lung following lung and stem-cell transplantation]. DER PATHOLOGE 2021; 42:17-24. [PMID: 33416936 DOI: 10.1007/s00292-020-00898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
Transplantation of solid organs and hematopoietic stem cells represents an important therapeutic option for a variety of end-stage pulmonary diseases, aggressive hematopoietic neoplasms, or severe immunodeficiencies. Although the overall survival following transplantation has generally improved over recent decades, long-time survival of lung and stem-cell transplant recipients is still alarmingly low with an average 5‑year survival rate of only 50-60%. Chronic allo-immunoreactions in general and pulmonary allo-immunoreactions with subsequent fibrosis in particular are major reasons for this poor outcome. Comparable patterns of fibrotic lung remodeling are observed following both lung and hematopoietic stem-cell transplantation. Besides the meanwhile well-established obliterative and functionally obstructive remodeling of the small airways - obliterative bronchiolitis - a specific restrictive subform of fibrosis, namely alveolar fibroelastosis, has been identified. Despite their crucial impact on patient outcome, both entities can be very challenging to detect by conventional histopathological analysis. Their underlying mechanisms are considered overreaching aberrant repair attempts to acute lung injuries with overactivation of (myo-) fibroblasts and excessive and irreversible deposition of extracellular matrix. Of note, the underlying molecular mechanisms are widely divergent between these two morphological entities and are independent of the underlying clinical setting.Further comprehensive investigations of these fibrotic alterations are key to the development of much-needed predictive diagnostics and curative concepts, considering the high mortality of pulmonary fibrosis following transplantation.
Collapse
Affiliation(s)
- Christopher Werlein
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland
| | - Max Ackermann
- Institut für Pathologie und Molekularpathologie, Helios Universitätsklinikum Wuppertal, Universität Witten-Herdecke, Wuppertal, Deutschland.,Institut für Funktionelle und Klinische Anatomie, Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Mainz, Deutschland
| | - Thia Leandra Hoffmann
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland
| | - Florian Laenger
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Standort Hannover, Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
| | - Danny Jonigk
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Standort Hannover, Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland.
| |
Collapse
|
24
|
Arjuna A, Olson MT, Walia R, Bremner RM, Smith MA, Mohanakumar T. An update on current treatment strategies for managing bronchiolitis obliterans syndrome after lung transplantation. Expert Rev Respir Med 2020; 15:339-350. [PMID: 33054424 DOI: 10.1080/17476348.2021.1835475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Bronchiolitis obliterans syndrome (BOS), a subtype of chronic lung allograft dysfunction, is quite common, with up to half of all lung recipients developing BOS within 5 years of transplantation. Preventive efforts are aimed at alleviating known risk factors of BOS development, while the primary goal of treatment is to delay the irreversible, fibrotic airway changes, and progressive loss of lung function. AREAS COVERED This narrative review will briefly discuss the updated definition, clinical presentation, pathogenesis, risk factors, and survival after BOS while paying particular attention to the salient evidence for optimal preventive strategies and treatments based on investigations in the modern era. EXPERT OPINION Future translational research focused on further characterizing the complex interplay between immune and nonimmune mechanisms mediating chronic lung rejection is the first step toward mitigating risk of allograft injury, improving early disease detection with noninvasive biomarkers, and ultimately, developing an effective, targeted therapy that can extend the life of the lung allograft.
Collapse
Affiliation(s)
- Ashwini Arjuna
- St. Joseph's Hospital and Medical Center, Norton Thoracic Institute, Phoenix, AZ, USA
| | - Michael T Olson
- St. Joseph's Hospital and Medical Center, Norton Thoracic Institute, Phoenix, AZ, USA.,Phoenix Campus, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Rajat Walia
- St. Joseph's Hospital and Medical Center, Norton Thoracic Institute, Phoenix, AZ, USA
| | - Ross M Bremner
- St. Joseph's Hospital and Medical Center, Norton Thoracic Institute, Phoenix, AZ, USA
| | - Michael A Smith
- St. Joseph's Hospital and Medical Center, Norton Thoracic Institute, Phoenix, AZ, USA
| | | |
Collapse
|