1
|
Ye D, Liu Q, Zhang C, Dai E, Fan J, Wu L. Relationship between immune cells and the development of chronic lung allograft dysfunction. Int Immunopharmacol 2024; 137:112381. [PMID: 38865754 DOI: 10.1016/j.intimp.2024.112381] [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: 04/03/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
A major cause of death for lung transplant recipients (LTRs) is the advent of chronic lung allograft dysfunction (CLAD), which has long plagued the long-term post-transplant prognosis and quality of survival of transplant patients. The intricacy of its pathophysiology and the irreversibility of its illness process present major obstacles to the clinical availability of medications. Immunotherapeutic medications are available, but they only aim to slow down the course of CLAD rather than having any therapeutic impact on the disease's development. For this reason, understanding the pathophysiology of CLAD is essential for both disease prevention and proven treatment. The immunological response in particular, in relation to chronic lung allograft dysfunction, has received a great deal of interest recently. Innate immune cells like natural killer cells, eosinophils, neutrophils, and mononuclear macrophages, as well as adaptive immunity cells like T and B cells, play crucial roles in this process through the release of chemokines and cytokines. The present review delves into changes and processes within the immune microenvironment, with a particular focus on the quantity, subtype, and characteristics of effector immune cells in the peripheral and transplanted lungs after lung transplantation. We incorporate and solidify the documented role of immune cells in the occurrence and development of CLAD with the advancements in recent years.
Collapse
Affiliation(s)
- Defeng Ye
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiongliang Liu
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengcheng Zhang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enci Dai
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiang Fan
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liang Wu
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Napoli C, Benincasa G, Fiorelli A, Strozziero MG, Costa D, Russo F, Grimaldi V, Hoetzenecker K. Lung transplantation: Current insights and outcomes. Transpl Immunol 2024; 85:102073. [PMID: 38889844 DOI: 10.1016/j.trim.2024.102073] [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: 11/16/2023] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Until now, the ability to predict or retard immune-mediated rejection events after lung transplantation is still limited due to the lack of specific biomarkers. The pressing need remains to early diagnose or predict the onset of chronic lung allograft dysfunction (CLAD) and its differential phenotypes that is the leading cause of death. Omics technologies (mainly genomics, epigenomics, and transcriptomics) combined with advanced bioinformatic platforms are clarifying the key immune-related molecular routes that trigger early and late events of lung allograft rejection supporting the biomarker discovery. The most promising biomarkers came from genomics. Both unregistered and NIH-registered clinical trials demonstrated that the increased percentage of donor-derived cell-free DNA in both plasma and bronchoalveolar lavage fluid showed a good diagnostic performance for clinically silent acute rejection events and CLAD differential phenotypes. A further success arose from transcriptomics that led to development of Molecular Microscope® Diagnostic System (MMDx) to interpret the relationship between molecular signatures of lung biopsies and rejection events. Other immune-related biomarkers of rejection events may be exosomes, telomer length, DNA methylation, and histone-mediated neutrophil extracellular traps (NETs) but none of them entered in registered clinical trials. Here, we discuss novel and existing technologies for revealing new immune-mediated mechanisms underlying acute and chronic rejection events, with a particular focus on emerging biomarkers for improving precision medicine of lung transplantation field.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Alfonso Fiorelli
- Thoracic Surgery Unit, Department of Translation Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | | | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | | | - Vincenzo Grimaldi
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
3
|
Hiho SJ, Levvey BJ, Diviney MB, Snell GI, Sullivan LC, Westall GP. Comparison of human leukocyte antigen immunologic risk stratification methods in lung transplantation. Am J Transplant 2024; 24:827-838. [PMID: 37981213 DOI: 10.1016/j.ajt.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
Outcomes after lung transplantation (LTx) remain poor, despite advances in sequencing technology and development of algorithms defining immunologic compatibility. Presently, there is no consensus regarding the best approach to define human leukocyte antigen (HLA) compatibility in LTx. In this study, we compared 5 different HLA compatibility tools in a high-resolution HLA-typed, clinically characterized cohort, to determine which approach predicts outcomes after LTx. In this retrospective single-center study, 277 donor-recipient transplant pairs were HLA-typed using next generation sequencing. HLA compatibility was defined using HLAMatchmaker, HLA epitope mismatch algorithm (HLA-EMMA), predicted indirectly recognizable HLA epitopes (PIRCHE), electrostatic mismatch score (EMS), and amino acid mismatches (AAMMs). Associations with HLA mismatching and survival, chronic lung allograft dysfunction (CLAD), and anti-HLA donor-specific antibody (DSA) were calculated using adjusted Cox proportional modeling. Lower HLA class II mismatching was associated with improved survival as defined by HLAMatchmaker (P < .01), HLA-EMMA (P < .05), PIRCHE (P < .05), EMS (P < .001), and AAMM (P < .01). All approaches demonstrated that HLA-DRB1345 matching was associated with freedom from restrictive allograft syndrome and HLA-DQ matching with reduced DSA development. Reducing the level of HLA mismatching, in T cell or B cell epitopes, electrostatic differences, or amino acid, can improve outcomes after LTx and potentially guide immunosuppression strategies.
Collapse
Affiliation(s)
- Steven J Hiho
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia; Australian Red Cross LifeBlood, Victorian Transplantation and Immunogenetics Service, Melbourne, Victoria, Australia.
| | - Bronwyn J Levvey
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Mary B Diviney
- Australian Red Cross LifeBlood, Victorian Transplantation and Immunogenetics Service, Melbourne, Victoria, Australia
| | - Gregory I Snell
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Lucy C Sullivan
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Australian Red Cross LifeBlood, South Australian Transplantation and Immunogenetics Service, Adelaide, South Australia, Australia
| | - Glen P Westall
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Tissot A, Durand E, Goronflot T, Coiffard B, Renaud-Picard B, Roux A, Demant X, Mornex JF, Falque L, Salpin M, Le Pavec J, Villeneuve T, Boussaud V, Knoop C, Magnan A, Lair D, Berthelot L, Danger R, Brouard S. Blood MMP-9 measured at 2 years after lung transplantation as a prognostic biomarker of chronic lung allograft dysfunction. Respir Res 2024; 25:88. [PMID: 38336710 PMCID: PMC10858575 DOI: 10.1186/s12931-024-02707-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Long-term outcomes of lung transplantation (LTx) remain hampered by chronic lung allograft dysfunction (CLAD). Matrix metalloproteinase 9 (MMP-9) is a secretory endopeptidase identified as a key mediator in fibrosis processes associated with CLAD. The objective of this study was to investigate whether plasma MMP9 levels may be prognostic of CLAD development. METHODS Participants were selected from the Cohort in Lung Transplantation (COLT) for which a biocollection was associated. We considered two time points, year 1 (Y1) and year 2 (Y2) post-transplantation, for plasma MMP-9 measurements. We analysed stable recipients at those time points, comparing those who would develop a CLAD within the 2 years following the measurement to those who would remain stable 2 years after. RESULTS MMP-9 levels at Y1 were not significantly different between the CLAD and stable groups (230 ng/ml vs. 160 ng/ml, p = 0.4). For the Y2 analysis, 129 recipients were included, of whom 50 developed CLAD within 2 years and 79 remained stable within 2 years. MMP-9 plasma median concentrations were higher in recipients who then developed CLAD than in the stable group (230 ng/ml vs. 118 ng/ml, p = 0.003). In the multivariate analysis, the Y2 MMP-9 level was independently associated with CLAD, with an average increase of 150 ng/ml (95% CI [0-253], p = 0.05) compared to that in the stable group. The Y2 ROC curve revealed a discriminating capacity of blood MMP-9 with an area under the curve of 66%. CONCLUSION Plasmatic MMP-9 levels measured 2 years after lung transplantation have prognostic value for CLAD.
Collapse
Affiliation(s)
- Adrien Tissot
- CHU Nantes, INSERM, Service de Pneumologie, l'institut du thorax, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes Université, 44093, Nantes, France.
| | - Eugénie Durand
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Thomas Goronflot
- CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, Nantes Université, Nantes, France
| | - Benjamin Coiffard
- Department of Respiratory Medicine and Lung Transplantation, APHM, Hôpital Nord, Aix Marseille Univ, Marseille, France
| | - Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, Inserm UMR 1260, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - Xavier Demant
- Service de Pneumologie, Centre Hospitalier Universitaire de Bordeaux, Pessac, France
| | - Jean-François Mornex
- Université Lyon 1, PSL, EPHE, INRAE, IVPC, Hospices Civils de Lyon, groupement hospitalier est, service de pneumologie, Orphalung, RESPIFIL, Université de Lyon, Lyon, France
| | - Loïc Falque
- Service Hospitalier Universitaire de Pneumologie et Physiologie, CHU Grenoble Alpes, Pôle Thorax et Vaisseaux, Grenoble, France
| | - Mathilde Salpin
- APHP Nord-Université Paris Cité, Hôpital Bichat, Service de Pneumologie B et Transplantation Pulmonaire, PHERE UMRS 1152, Université Paris Cité, Paris, France
| | - Jérôme Le Pavec
- Service de Pneumologie et Transplantation Pulmonaire, Groupe hospitalier Marie-Lannelongue -Saint Joseph, Le Plessis-Robinson, Université Paris-Saclay, UMR_S 999, INSERM, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Thomas Villeneuve
- CHU Toulouse, Service de Pneumologie, Université Toulouse III-Paul Sabatier, Toulouse, France
| | | | | | - Antoine Magnan
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - David Lair
- CHU Nantes, Nantes Université, Institut du Thorax, Lung O2, Nantes, France
| | - Laureline Berthelot
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Richard Danger
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Sophie Brouard
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| |
Collapse
|
5
|
Biomarkers for Chronic Lung Allograft Dysfunction: Ready for Prime Time? Transplantation 2023; 107:341-350. [PMID: 35980878 PMCID: PMC9875844 DOI: 10.1097/tp.0000000000004270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle impairing lung transplant outcome. Parallel to the better clinical identification and characterization of CLAD and CLAD phenotypes, there is an increasing urge to find adequate biomarkers that could assist in the earlier detection and differential diagnosis of CLAD phenotypes, as well as disease prognostication. The current status and state-of-the-art of biomarker research in CLAD will be discussed with a particular focus on radiological biomarkers or biomarkers found in peripheral tissue, bronchoalveolar lavage' and circulating blood' in which significant progress has been made over the last years. Ultimately, although a growing number of biomarkers are currently being embedded in the follow-up of lung transplant patients, it is clear that one size does not fit all. The future of biomarker research probably lies in the rigorous combination of clinical information with findings in tissue, bronchoalveolar lavage' or blood. Only by doing so, the ultimate goal of biomarker research can be achieved, which is the earlier identification of CLAD before its clinical manifestation. This is desperately needed to improve the prognosis of patients with CLAD after lung transplantation.
Collapse
|
6
|
Markers of Bronchiolitis Obliterans Syndrome after Lung Transplant: Between Old Knowledge and Future Perspective. Biomedicines 2022; 10:biomedicines10123277. [PMID: 36552035 PMCID: PMC9775233 DOI: 10.3390/biomedicines10123277] [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: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the most common form of CLAD and is characterized by airflow limitation and an obstructive spirometric pattern without high-resolution computed tomography (HRCT) evidence of parenchymal opacities. Computed tomography and microCT analysis show abundant small airway obstruction, starting from the fifth generation of airway branching and affecting up to 40-70% of airways. The pathogenesis of BOS remains unclear. It is a multifactorial syndrome that leads to pathological tissue changes and clinical manifestations. Because BOS is associated with the worst long-term survival in LTx patients, many studies are focused on the early identification of BOS. Markers may be useful for diagnosis and for understanding the molecular and immunological mechanisms involved in the onset of BOS. Diagnostic and predictive markers of BOS have also been investigated in various biological materials, such as blood, BAL, lung tissue and extracellular vesicles. The aim of this review was to evaluate the scientific literature on markers of BOS after lung transplant. We performed a systematic review to find all available data on potential prognostic and diagnostic markers of BOS.
Collapse
|
7
|
Bos S, Milross L, Filby AJ, Vos R, Fisher AJ. Immune processes in the pathogenesis of chronic lung allograft dysfunction: identifying the missing pieces of the puzzle. Eur Respir Rev 2022; 31:31/165/220060. [PMID: 35896274 DOI: 10.1183/16000617.0060-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/19/2022] [Indexed: 11/05/2022] Open
Abstract
Lung transplantation is the optimal treatment for selected patients with end-stage chronic lung diseases. However, chronic lung allograft dysfunction remains the leading obstacle to improved long-term outcomes. Traditionally, lung allograft rejection has been considered primarily as a manifestation of cellular immune responses. However, in reality, an array of complex, interacting and multifactorial mechanisms contribute to its emergence. Alloimmune-dependent mechanisms, including T-cell-mediated rejection and antibody-mediated rejection, as well as non-alloimmune injuries, have been implicated. Moreover, a role has emerged for autoimmune responses to lung self-antigens in the development of chronic graft injury. The aim of this review is to summarise the immune processes involved in the pathogenesis of chronic lung allograft dysfunction, with advanced insights into the role of innate immune pathways and crosstalk between innate and adaptive immunity, and to identify gaps in current knowledge.
Collapse
Affiliation(s)
- Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Luke Milross
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Andrew J Filby
- Flow Cytometry Core and Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Robin Vos
- Dept of CHROMETA, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.,University Hospitals Leuven, Dept of Respiratory Diseases, Leuven, Belgium
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK .,Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
8
|
Smirnova NF, Riemondy K, Bueno M, Collins S, Suresh P, Wang X, Patel KN, Cool C, Königshoff M, Sharma NS, Eickelberg O. Single-cell transcriptome mapping identifies a local, innate B cell population driving chronic rejection after lung transplantation. JCI Insight 2022; 7:156648. [PMID: 36134664 PMCID: PMC9675462 DOI: 10.1172/jci.insight.156648] [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: 12/02/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the main reason for poor outcomes after lung transplantation (LTx). We and others have recently identified B cells as major contributors to BOS after LTx. The extent of B cell heterogeneity and the relative contributions of B cell subpopulations to BOS, however, remain unclear. Here, we provide a comprehensive analysis of cell population changes and their gene expression patterns during chronic rejection after orthotopic LTx in mice. Of 11 major cell types, Mzb1-expressing plasma cells (PCs) were the most prominently increased population in BOS lungs. These findings were validated in 2 different cohorts of human BOS after LTx. A Bhlhe41, Cxcr3, and Itgb1 triple-positive B cell subset, also expressing classical markers of the innate-like B-1 B cell population, served as the progenitor pool for Mzb1+ PCs. This subset accounted for the increase in IgG2c production within BOS lung grafts. A genetic lack of Igs decreased BOS severity after LTx. In summary, we provide a detailed analysis of cell population changes during BOS. IgG+ PCs and their progenitors — an innate B cell subpopulation — are the major source of local Ab production and a significant contributor to BOS after LTx.
Collapse
Affiliation(s)
- Natalia F Smirnova
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Institut des Maladies Métaboliques et Cardiovasculaires (I2MC) - INSERM U1297, University of Toulouse III, Toulouse, France
| | - Kent Riemondy
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Marta Bueno
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Susan Collins
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Pavan Suresh
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kapil N Patel
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida/Tampa General Hospital, Tampa, Florida, USA
| | - Carlyne Cool
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nirmal S Sharma
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida/Tampa General Hospital, Tampa, Florida, USA.,Division of Pulmonary & Critical Care, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Pulmonary graft-versus-host disease and chronic lung allograft dysfunction: two sides of the same coin? THE LANCET RESPIRATORY MEDICINE 2022; 10:796-810. [DOI: 10.1016/s2213-2600(22)00001-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022]
|
10
|
de Fays C, Carlier FM, Gohy S, Pilette C. Secretory Immunoglobulin A Immunity in Chronic Obstructive Respiratory Diseases. Cells 2022; 11:1324. [PMID: 35456002 PMCID: PMC9027823 DOI: 10.3390/cells11081324] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), asthma and cystic fibrosis (CF) are distinct respiratory diseases that share features such as the obstruction of small airways and disease flare-ups that are called exacerbations and are often caused by infections. Along the airway epithelium, immunoglobulin (Ig) A contributes to first line mucosal protection against inhaled particles and pathogens. Dimeric IgA produced by mucosal plasma cells is transported towards the apical pole of airway epithelial cells by the polymeric Ig receptor (pIgR), where it is released as secretory IgA. Secretory IgA mediates immune exclusion and promotes the clearance of pathogens from the airway surface by inhibiting their adherence to the epithelium. In this review, we summarize the current knowledge regarding alterations of the IgA/pIgR system observed in those major obstructive airway diseases and discuss their implication for disease pathogenesis.
Collapse
Affiliation(s)
- Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
| | - François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, CHU UCL Namur, Site Mont-Godinne, 5530 Yvoir, Belgium
- Lung Transplant Centre, CHU UCL Namur, Site Mont-Godinne, 5530 Yvoir, Belgium
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (C.d.F.); (F.M.C.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
11
|
Silva TD, Voisey J, Hopkins P, Apte S, Chambers D, O'Sullivan B. Markers of rejection of a lung allograft: state of the art. Biomark Med 2022; 16:483-498. [PMID: 35315284 DOI: 10.2217/bmm-2021-1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) affects approximately 50% of all lung transplant recipients by 5 post-operative years and is the leading cause of death in lung transplant recipients. Early CLAD diagnosis or ideally prediction of CLAD is essential to enable early intervention before significant lung injury occurs. New technologies have emerged to facilitate biomarker discovery, including epigenetic modification and single-cell RNA sequencing. This review examines new and existing technologies for biomarker discovery and the current state of research on biomarkers for identifying lung transplant rejection.
Collapse
Affiliation(s)
- Tharushi de Silva
- School of Biomedical Sciences, Centre for Genomics & Personalised Heath, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, 4032, Brisbane, Queensland, Australia
| | - Joanne Voisey
- School of Biomedical Sciences, Centre for Genomics & Personalised Heath, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Peter Hopkins
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, 4032, Brisbane, Queensland, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, The University of Queensland, 4032, Brisbane, Queensland, Australia
| | - Simon Apte
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, 4032, Brisbane, Queensland, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, The University of Queensland, 4032, Brisbane, Queensland, Australia
| | - Daniel Chambers
- School of Biomedical Sciences, Centre for Genomics & Personalised Heath, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, 4032, Brisbane, Queensland, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, The University of Queensland, 4032, Brisbane, Queensland, Australia
| | - Brendan O'Sullivan
- School of Biomedical Sciences, Centre for Genomics & Personalised Heath, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, 4032, Brisbane, Queensland, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, The University of Queensland, 4032, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Bos S, Filby AJ, Vos R, Fisher AJ. Effector immune cells in Chronic Lung Allograft Dysfunction: a Systematic Review. Immunology 2022; 166:17-37. [PMID: 35137398 PMCID: PMC9426626 DOI: 10.1111/imm.13458] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/13/2022] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) remains the major barrier to long‐term survival after lung transplantation and improved insight into its underlying immunological mechanisms is critical to better understand the disease and to identify treatment targets. We systematically searched the electronic databases of PubMed and EMBASE for original research publications, published between January 2000 and April 2021, to comprehensively assess current evidence on effector immune cells in lung tissue and bronchoalveolar lavage fluid from lung transplant recipients with CLAD. Literature search revealed 1351 articles, 76 of which met the criteria for inclusion in our analysis. Our results illustrate significant complexity in both innate and adaptive immune cell responses in CLAD, along with presence of numerous immune cell products, including cytokines, chemokines and proteases associated with tissue remodelling. A clear link between neutrophils and eosinophils and CLAD incidence has been seen, in which eosinophils more specifically predisposed to restrictive allograft syndrome. The presence of cytotoxic and T‐helper cells in CLAD pathogenesis is well‐documented, although it is challenging to draw conclusions about their role in tissue processes from predominantly bronchoalveolar lavage data. In restrictive allograft syndrome, a more prominent humoral immune involvement with increased B cells, immunoglobulins and complement deposition is seen. Our evaluation of published studies over the last 20 years summarizes the complex multifactorial immunopathology of CLAD onset and progression. It highlights the phenotype of several key effector immune cells involved in CLAD pathogenesis, as well as the paucity of single cell resolution spatial studies in lung tissue from patients with CLAD.
Collapse
Affiliation(s)
- Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom.,Institute of Transplantation, The Newcastle Upon Tyne Hospital NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Andrew J Filby
- Flow Cytometry Core and Innovation, Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Robin Vos
- Department of CHROMETA, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.,University Hospitals Leuven, Dept. of Respiratory Diseases, Leuven, Belgium
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom.,Institute of Transplantation, The Newcastle Upon Tyne Hospital NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
13
|
Divithotawela C, Pham A, Bell PT, Ledger EL, Tan M, Yerkovich S, Grant M, Hopkins PM, Wells TJ, Chambers DC. Inferior outcomes in lung transplant recipients with serum Pseudomonas aeruginosa specific cloaking antibodies. J Heart Lung Transplant 2021; 40:951-959. [PMID: 34226118 DOI: 10.1016/j.healun.2021.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/21/2021] [Accepted: 05/24/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Chronic Lung Allograft Dysfunction (CLAD) limits long-term survival following lung transplantation. Colonization of the allograft by Pseudomonas aeruginosa is associated with an increased risk of CLAD and inferior overall survival. Recent experimental data suggests that 'cloaking' antibodies targeting the O-antigen of the P. aeruginosa lipopolysaccharide cell wall (cAbs) attenuate complement-mediated bacteriolysis in suppurative lung disease. METHODS In this retrospective cohort analysis of 123 lung transplant recipients, we evaluated the prevalence, risk factors and clinical impact of serum cAbs following transplantation. RESULTS cAbs were detected in the sera of 40.7% of lung transplant recipients. Cystic fibrosis and younger age were associated with increased risk of serum cAbs (CF diagnosis, OR 6.62, 95% CI 2.83-15.46, p < .001; age at transplant, OR 0.69, 95% CI 0.59-0.81, p < .001). Serum cAbs and CMV mismatch were both independently associated with increased risk of CLAD (cAb, HR 4.34, 95% CI 1.91-9.83, p < .001; CMV mismatch (D+/R-), HR 5.40, 95% CI 2.36-12.32, p < .001) and all-cause mortality (cAb, HR 2.75, 95% CI 1.27-5.95, p = .010, CMV mismatch, HR 3.53, 95% CI 1.62-7.70, p = .002) in multivariable regression analyses. CONCLUSIONS Taken together, these findings suggest a potential role for 'cloaking' antibodies targeting P. aeruginosa LPS O-antigen in the immunopathogenesis of CLAD.
Collapse
Affiliation(s)
| | - Amy Pham
- The University of Queensland, Diamantina Institute, The University of Queensland, Wooloongabba, Australia
| | - Peter T Bell
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Australia; School of Medicine, The University of Queensland, Brisbane, Australia
| | - Emma L Ledger
- The University of Queensland, Diamantina Institute, The University of Queensland, Wooloongabba, Australia
| | - Maxine Tan
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Australia
| | | | - Michelle Grant
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Australia
| | - Peter M Hopkins
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Australia; School of Medicine, The University of Queensland, Brisbane, Australia
| | - Timothy J Wells
- The University of Queensland, Diamantina Institute, The University of Queensland, Wooloongabba, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Daniel C Chambers
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Australia; School of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
14
|
Amubieya O, Ramsey A, DerHovanessian A, Fishbein GA, Lynch JP, Belperio JA, Weigt SS. Chronic Lung Allograft Dysfunction: Evolving Concepts and Therapies. Semin Respir Crit Care Med 2021; 42:392-410. [PMID: 34030202 DOI: 10.1055/s-0041-1729175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary factor that limits long-term survival after lung transplantation is chronic lung allograft dysfunction (CLAD). CLAD also impairs quality of life and increases the costs of medical care. Our understanding of CLAD continues to evolve. Consensus definitions of CLAD and the major CLAD phenotypes were recently updated and clarified, but it remains to be seen whether the current definitions will lead to advances in management or impact care. Understanding the potential differences in pathogenesis for each CLAD phenotype may lead to novel therapeutic strategies, including precision medicine. Recognition of CLAD risk factors may lead to earlier interventions to mitigate risk, or to avoid risk factors all together, to prevent the development of CLAD. Unfortunately, currently available therapies for CLAD are usually not effective. However, novel therapeutics aimed at both prevention and treatment are currently under investigation. We provide an overview of the updates to CLAD-related terminology, clinical phenotypes and their diagnosis, natural history, pathogenesis, and potential strategies to treat and prevent CLAD.
Collapse
Affiliation(s)
- Olawale Amubieya
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Allison Ramsey
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ariss DerHovanessian
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gregory A Fishbein
- Department of Pathology, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Joseph P Lynch
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - John A Belperio
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - S Samuel Weigt
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
15
|
Takeshita M, Suzuki K, Nakazawa M, Kamata H, Ishii M, Oyamada Y, Oshima H, Takeuchi T. Antigen-driven autoantibody production in lungs of interstitial lung disease with autoimmune disease. J Autoimmun 2021; 121:102661. [PMID: 34034155 DOI: 10.1016/j.jaut.2021.102661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022]
Abstract
Interstitial lung disease (ILD) sometimes becomes a life-threatening complication of systemic autoimmune diseases; however, little is known about the immune response in lung lesions. We aimed to investigate humoural immunity in ILD associated with rheumatoid arthritis (RA), sjögren's syndrome (SjS), and mixed connective tissue disease (MCTD), using bronchoalveolar fluid (BALF) and serum samples from 15 patients with autoimmune disease associated-ILD. We first showed that BALF contained higher titers of disease-related autoantibodies than serum, suggesting the possibility of autoantibody production in lungs. Next, we produced 326 monoclonal antibodies from antibody-secreting cells in BALF, and the reactivity and their revertants, in which somatic hypermutations were reverted to germline, were analyzed. Among 123 antibodies from RA-ILD, 16 disease-related antibodies (anti-modified protein antibodies and rheumatoid factors) were identified, of which one antibody had both properties. The revertant antibodies changed their target modification in a complicated manner, suggesting that the antibodies were selected against various modifications in lungs. Among 146 antibodies from SjS-ILD and/or MCTD-ILD, seven anti-SSA/Ro60 antibodies and 15 anti-RNP antibodies were identified. Some of the anti-RNP antibodies recognized multiple RNP constituent proteins simultaneously, indicating that epitope spreading may progress in lungs. Our results revealed the existence of an active autoimmunity in the lungs of autoimmune disease associated-ILD.
Collapse
Affiliation(s)
- Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Maho Nakazawa
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshitaka Oyamada
- Department of Respiratory Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Hisaji Oshima
- Department of Connective Tissue Diseases, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Heigl T, Saez-Gimenez B, Van Herck A, Kaes J, Sacreas A, Beeckmans H, Ambrocio GPL, Kwakkel-Van Erp H, Ordies S, Vanstapel A, Verleden SE, Neyrinck AP, Ceulemans LJ, Van Raemdonck DE, Verbeken E, Verleden GM, Vos R, Vanaudenaerde B. Free Airway C4d after Lung Transplantation - A Quantitative Analysis of Bronchoalveolar Lavage Fluid. Transpl Immunol 2020; 64:101352. [PMID: 33217540 DOI: 10.1016/j.trim.2020.101352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/07/2020] [Accepted: 11/07/2020] [Indexed: 01/23/2023]
Abstract
In recent years, the utility of vascular complement factor 4d (C4d) deposition as diagnostic tool for antibody mediated rejection (AMR) after lung transplantation, has become a controversial issue. We aimed to pinpoint the problematic nature of C4d as biomarker with a simple experiment. We quantified C4d in broncho-alveolar lavage (BAL) of lung transplant patients with diverse post-transplant complications in 3 different settings of clinically clear cases of: 1/ chronic lung allograft dysfunction (CLAD); 2/ acute complications acute rejection (AR), lymphocytic bronchiolitis (LB), antibody-mediated rejection (AMR) and respiratory infection (INF); 3/ patients with parallel C4d immunostaining and Anti-HLA. All groups were compared to BAL of stable patients. C4d was measured via standard ELISA. C4d was increased in CLAD, predominantly in RAS (p = 0.0026) but not in BOS (p = 0.89). C4d was increased in all acute events, AR (p = 0.0025), LB (p < 0.0001), AMR (p = 0.0034), infections (p < 0.0001). In patients with parallel C4d immunostaining and serum HLA antibodies, C4d was increased in C4d-/HLA- (p = 0.0011); C4d-/HLA+ (p = 0.013); HLA+/C4d + (p = 0.0081). A correlation of systemic C-reactive protein (CRP) with C4d was found in all patients (r = 0.49; p < 0.0001). We hypothesize that free C4d in BAL may only be representative of a general immune response in the transplanted lung.
Collapse
Affiliation(s)
- Tobias Heigl
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Berta Saez-Gimenez
- Department of Pneumology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Anke Van Herck
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Hanne Beeckmans
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Gene P L Ambrocio
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Hanneke Kwakkel-Van Erp
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium; Department of Pneumology, University of Antwerp, Antwerp, Belgium
| | - Sofie Ordies
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Erik Verbeken
- Translational Cell and Tissue Research, KU Leuven and UZ Gasthuisberg, Leuven, Belgium
| | - Geert M Verleden
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium
| | - Bart Vanaudenaerde
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), UZ/KU Leuven, Leuven, Belgium.
| |
Collapse
|
17
|
Yoshiyasu N, Sato M. Chronic lung allograft dysfunction post-lung transplantation: The era of bronchiolitis obliterans syndrome and restrictive allograft syndrome. World J Transplant 2020; 10:104-116. [PMID: 32864356 PMCID: PMC7428788 DOI: 10.5500/wjt.v10.i5.104] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 02/05/2023] Open
Abstract
Chronic lung allograft dysfunction (CLAD) following lung transplantation limits long-term survival considerably. The main reason for this is a lack of knowledge regarding the pathological condition and the establishment of treatment. The consensus statement from the International Society for Heart and Lung Transplantation on CLAD in 2019 classified CLAD into two main phenotypes: Bronchiolitis obliterans syndrome and restrictive allograft syndrome. Along with this clear classification, further exploration of the mechanisms and the development of appropriate prevention and treatment strategies for each phenotype are desired. In this review, we summarize the new definition of CLAD and update and summarize the existing knowledge on the underlying mechanisms of bronchiolitis obliterans syndrome and restrictive allograft syndrome, which have been elucidated from clinicopathological observations and animal experiments worldwide.
Collapse
Affiliation(s)
- Nobuyuki Yoshiyasu
- Department of Thoracic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| |
Collapse
|
18
|
Combs MP, Xia M, Wheeler DS, Belloli EA, Walker NM, Braeuer RR, Lyu DM, Murray S, Lama VN. Fibroproliferation in chronic lung allograft dysfunction: Association of mesenchymal cells in bronchoalveolar lavage with phenotypes and survival. J Heart Lung Transplant 2020; 39:815-823. [PMID: 32360292 DOI: 10.1016/j.healun.2020.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD), the primary cause of poor outcome after lung transplantation, arises from fibrotic remodeling of the allograft and presents as diverse clinical phenotypes with variable courses. Here, we investigate whether bronchoalveolar lavage (BAL) mesenchymal cell activity at CLAD onset can inform regarding disease phenotype, progression, and survival. METHODS Mesenchymal cell colony-forming units (CFUs) were measured in BAL obtained at CLAD onset (n = 77) and CLAD-free time post-transplant matched controls (n = 77). CFU counts were compared using Wilcoxon's rank-sum test. Cox proportional hazards and restricted means models were utilized to investigate post-CLAD survival. RESULTS Higher mesenchymal CFU counts were noted in BAL at the time of CLAD onset than in CLAD-free controls. Patients with restrictive allograft syndrome had higher BAL mesenchymal CFU count at CLAD onset than patients with bronchiolitis obliterans syndrome (p = 0.011). Patients with high mesenchymal CFU counts (≥10) at CLAD onset had worse outcomes than those with low (<10) CFU counts, with shorter average survival (2.64 years vs 4.25 years; p = 0.027) and shorter progression-free survival, defined as time to developing either CLAD Stage 3 or death (0.97 years vs 2.70 years; p < 0.001). High CFU count remained predictive of decreased overall survival and progression-free survival after accounting for the CLAD phenotype and other clinical factors in multivariable analysis. CONCLUSIONS Fulminant fibroproliferation with higher mesenchymal CFU counts in BAL is noted in restrictive allograft syndrome and is independently associated with poor survival after CLAD onset.
Collapse
Affiliation(s)
- Michael P Combs
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Meng Xia
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - David S Wheeler
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth A Belloli
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Natalie M Walker
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Russell R Braeuer
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Dennis M Lyu
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Susan Murray
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Vibha N Lama
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
19
|
Abstract
Introduction: Lung transplantation remains an important treatment for patients with end stage lung disease. Chronic lung allograft dysfunction (CLAD) remains the greatest limiting factor for long term survival. As the diagnosis of CLAD is based on pulmonary function tests, significant lung injury is required before a diagnosis is feasible, likely when irreversible damage has already occurred. Therefore, research is ongoing for early CLAD recognition, with biomarkers making up a substantial amount of this research.Areas covered: The purpose of this review is to describe available biomarkers, focusing on those which aid in predicting CLAD and distinguishing between different CLAD phenotypes. We describe biomarkers presenting in bronchial alveolar lavage (BAL) as well as circulating in peripheral blood, both of which offer an appealing alternative to lung biopsy.Expert opinion: Development of CLAD involves complex, multiple immune and nonimmune mechanisms. Therefore, evaluation of potential CLAD biomarkers serves a dual purpose: clinically, the goal remains early detection and identification of patients at increased risk. Simultaneously, biomarkers offer insight into the different mechanisms involved in the pathophysiology of CLAD, leading to the development of possible interventions. The ultimate goal is the development of both preventive and early intervention strategies for CLAD to improve the overall survival of our lung transplant recipients.
Collapse
Affiliation(s)
- Osnat Shtraichman
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Pulmonary institute, Rabin Medical Center, Petach Tikva, Israel; Sackler School of Medicine, Tel Aviv, Israel
| | - Joshua M Diamond
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Sato M. Bronchiolitis obliterans syndrome and restrictive allograft syndrome after lung transplantation: why are there two distinct forms of chronic lung allograft dysfunction? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:418. [PMID: 32355862 PMCID: PMC7186721 DOI: 10.21037/atm.2020.02.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bronchiolitis obliterans syndrome (BOS) had been considered to be the representative form of chronic rejection or chronic lung allograft dysfunction (CLAD) after lung transplantation. In BOS, small airways are affected by chronic inflammation and obliterative fibrosis, whereas peripheral lung tissue remains relatively intact. However, recognition of another form of CLAD involving multiple tissue compartments in the lung, termed restrictive allograft syndrome (RAS), raised a fundamental question: why there are two phenotypes of CLAD? Increasing clinical and experimental data suggest that RAS may be a prototype of chronic rejection after lung transplantation involving both cellular and antibody-mediated alloimmune responses. Some cases of RAS are also induced by fulminant general inflammation in lung allografts. However, BOS involves alloimmune responses and the airway-centered disease process can be explained by multiple mechanisms such as external alloimmune-independent stimuli (such as infection, aspiration and air pollution), exposure of airway-specific autoantigens and airway ischemia. Localization of immune responses in different anatomical compartments in different phenotypes of CLAD might be associated with lymphoid neogenesis or the de novo formation of lymphoid tissue in lung allografts. Better understanding of distinct mechanisms of BOS and RAS will facilitate the development of effective preventive and therapeutic strategies of CLAD.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Sacreas A, Taupin JL, Emonds MP, Daniëls L, Van Raemdonck DE, Vos R, Verleden GM, Vanaudenaerde BM, Roux A, Verleden SE. Intragraft donor-specific anti-HLA antibodies in phenotypes of chronic lung allograft dysfunction. Eur Respir J 2019; 54:13993003.00847-2019. [PMID: 31439680 DOI: 10.1183/13993003.00847-2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/31/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Circulating anti-human leukocyte antigen (HLA) serum donor-specific antibodies (sDSAs) increase the risk of chronic lung allograft dysfunction (CLAD) and mortality. Discrepancies between serological and pathological/clinical findings are common. Therefore, we aimed to assess the presence of tissue-bound graft DSAs (gDSAs) in CLAD explant tissue compared with sDSAs. METHODS Tissue cores, obtained from explant lungs of unused donors (n=10) and patients with bronchiolitis obliterans syndrome (BOS; n=18) and restrictive allograft syndrome (RAS; n=18), were scanned with micro-computed tomography before elution of antibodies. Total IgG levels were measured via ELISA. Anti-HLA class I and II IgG gDSAs were identified using Luminex single antigen beads and compared with DSAs found in serum samples. RESULTS Overall, mean fluorescence intensity was higher in RAS eluates compared with BOS and controls (p<0.0001). In BOS, two patients were sDSA+/gDSA+ and two patients were sDSA-/gDSA+. In RAS, four patients were sDSA+/gDSA+, one patient was sDSA+/gDSA- and five patients were sDSA-/gDSA+. Serum and graft results combined, DSAs were more prevalent in RAS compared with BOS (56% versus 22%; p=0.04). There was spatial variability in gDSA detection in one BOS patient and three RAS patients, who were all sDSA-. Total graft IgG levels were higher in RAS than BOS (p<0.0001) and in gDSA+ versus gDSA- (p=0.0008), but not in sDSA+ versus sDSA- (p=0.33). In RAS, total IgG levels correlated with fibrosis (r= -0.39; p=0.02). CONCLUSIONS This study underlines the potential of gDSA assessment as complementary information to sDSA findings. The relevance and applications of gDSAs need further investigation.
Collapse
Affiliation(s)
- Annelore Sacreas
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Jean-Luc Taupin
- Laboratoire d'Immunologie et Histocompatibilité, Saint-Louis Hospital, Paris, France
| | - Marie-Paule Emonds
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium.,Dept of Immunology and Microbiology, KU Leuven, Leuven, Belgium
| | - Liesbeth Daniëls
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Dept of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Antoine Roux
- Service de Transplantation Pulmonaire, Foch Hospital, Suresnes, France
| | - Stijn E Verleden
- Leuven Lung Transplant Group, Dept of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | | |
Collapse
|
22
|
Kardol-Hoefnagel T, Budding K, van de Graaf EA, van Setten J, van Rossum OA, Oudijk EJD, Otten HG. A Single Nucleotide C3 Polymorphism Associates With Clinical Outcome After Lung Transplantation. Front Immunol 2019; 10:2245. [PMID: 31616421 PMCID: PMC6775212 DOI: 10.3389/fimmu.2019.02245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Development of chronic rejection is still a severe problem and causes high mortality rates after lung transplantation (LTx). Complement activation is important in the development of acute rejection (AR) and bronchiolitis obliterans syndrome, with C3 as a key complement factor. Methods: We investigated a single nucleotide polymorphism (SNP) in the C3 gene (rs2230199) in relation to long-term outcome after LTx in 144 patient-donor pairs. In addition, we looked at local production of donor C3 by analyzing bronchoalveolar lavage fluid (BALF) of 6 LTx patients using isoelectric focusing (IEF). Results: We demonstrated the presence of C3 in BALF and showed that this is produced by the donor lung based on the genotype of SNP rs2230199. We also analyzed donor and patient SNP configurations and observed a significant association between the SNP configuration in patients and episodes of AR during 4-years follow-up. Survival analysis showed a lower AR-free survival in homozygous C3 slow patients (p = 0.005). Furthermore, we found a significant association between the SNP configuration in donors and BOS development. Patients receiving a graft from a donor with at least one C3 fast variant for rs2230199 had an inferior BOS-free survival (p = 0.044). Conclusions: In conclusion, our data indicate local C3 production by donor lung cells. In addition, a single C3 SNP present in recipients affects short-term outcome after LTx, while this SNP in donors has an opposite effect on long-term outcome after LTx. These results could contribute to an improved risk stratification after transplantation.
Collapse
Affiliation(s)
- Tineke Kardol-Hoefnagel
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kevin Budding
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Eduard A van de Graaf
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Oliver A van Rossum
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Erik-Jan D Oudijk
- Center of Interstitial Lung Diseases, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Henderikus G Otten
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
23
|
Li Y, Shu P, Tang L, Yang X, Fan J, Zhang X. FK506 combined with GM6001 prevents tracheal obliteration in a mouse model of heterotopic tracheal transplantation. Transpl Immunol 2019; 57:101244. [PMID: 31526865 DOI: 10.1016/j.trim.2019.101244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/08/2019] [Accepted: 09/13/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Obliterative bronchiolitis (OB) is the major complication limiting the long-term survival of allografts after lung transplantation. In this study, we investigated the effect of tacrolimus (FK506) combined with GM6001,a matrix metalloproteinase (MMP) inhibitor, on the formation of OB using a mouse heterotopic tracheal transplantation model. METHODS Syngeneic tracheal grafts were transplanted heterotopically from BALB/c mice to BALB/c mice. Allografts from C57BL/6 mice were transplanted to BALB/c mice. Isograft group, allograft group, allograft+FK506 group, allograft +GM6001 group and allograft+FK506 + GM6001 group was given respectively intraperitoneal injection of saline, saline, FK506, GM6001 and FK506 + GM6001 once a day. At 28 day after transplantation, OB incidence was determined by hematoxylin-eosin staining and the expressions of MMPs and cytokines were assessed using enzyme linked immunosorbent assay, immunohistochemical assays and western blot assay. RESULTS The tracheal occlusion rates of isograft group, allograft group, allograft+FK506 group, allograft+GM6001 group and allograft+FK506 + GM6001 group were 0, 74.1 ± 9.79%, 34.4 ± 6.04%, 40.3 ± 8.77% and 26.5 ± 5.73% respectively. There were significant differences between the latter two groups (P < .001). The serum MMP-8 and MMP-9 levels of allograft group were significantly higher than those of isograft group (P < .05) and had no significant decrease when treated by FK506. The serum MMP-8 and MMP-9 levels of allograft+FK506 + GM6001 group were significantly lower than those of allograft+FK506 group (P < .05). MMP-8 and MMP-9 protein expression in the grafts of allograft+FK506 + GM6001 group were lower than those of allograft+FK506 group verified by immunohistochemical staining and western blotting. CONCLUSION FK506 combined with GM6001 could alleviate tracheal obliteration in mouse heterotopic tracheal transplantation model, due to its inhibitory effect on MMPs.
Collapse
Affiliation(s)
- Yiqian Li
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Ping Shu
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Liang Tang
- Department of central Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Xiaojun Yang
- Department of central Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Junwei Fan
- Department of general Surgery, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China.
| | - Xiaoqing Zhang
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China.
| |
Collapse
|
24
|
Tissot A, Danger R, Claustre J, Magnan A, Brouard S. Early Identification of Chronic Lung Allograft Dysfunction: The Need of Biomarkers. Front Immunol 2019; 10:1681. [PMID: 31379869 PMCID: PMC6650588 DOI: 10.3389/fimmu.2019.01681] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/04/2019] [Indexed: 01/12/2023] Open
Abstract
A growing number of patients with end-stage lung disease have benefited from lung transplantation (LT). Improvements in organ procurement, surgical techniques and intensive care management have greatly increased short-term graft survival. However, long-term outcomes remain limited, mainly due to the onset of chronic lung allograft dysfunction (CLAD), whose diagnosis is based on permanent loss of lung function after the development of irreversible lung lesions. CLAD is associated with high mortality and morbidity, and its exact physiopathology is still only partially understood. Many researchers and clinicians have searched for CLAD biomarkers to improve diagnosis, to refine the phenotypes associated with differential prognosis and to identify early biological processes that lead to CLAD to enable an early intervention that could modify the inevitable degradation of respiratory function. Donor-specific antibodies are currently the only biomarkers used in routine clinical practice, and their significance for accurately predicting CLAD is still debated. We describe here significant studies that have highlighted potential candidates for reliable and non-invasive biomarkers of CLAD in the fields of imaging and functional monitoring, humoral immunity, cell-mediated immunity, allograft injury, airway remodeling and gene expression. Such biomarkers would improve CLAD prediction and allow differential LT management regarding CLAD risk.
Collapse
Affiliation(s)
- Adrien Tissot
- Centre de Recherche en Transplantation et Immunologie (CRTI), INSERM, Université de Nantes, Nantes, France.,Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France
| | - Richard Danger
- Centre de Recherche en Transplantation et Immunologie (CRTI), INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Johanna Claustre
- Centre de Recherche en Transplantation et Immunologie (CRTI), INSERM, Université de Nantes, Nantes, France.,Service Hospitalo-Universitaire de Pneumologie - Physiologie, CHU Grenoble Alpes, Grenoble, France
| | - Antoine Magnan
- Centre de Recherche en Transplantation et Immunologie (CRTI), INSERM, Université de Nantes, Nantes, France.,Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,UMR S 1087 CNRS UMR 6291, Institut du Thorax, CHU Nantes, Université de Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie (CRTI), INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
25
|
Vos R, Eynde RV, Ruttens D, Verleden SE, Vanaudenaerde BM, Dupont LJ, Yserbyt J, Verbeken EK, Neyrinck AP, Van Raemdonck DE, Verleden GM. Montelukast in chronic lung allograft dysfunction after lung transplantation. J Heart Lung Transplant 2019; 38:516-527. [DOI: 10.1016/j.healun.2018.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
|
26
|
Parker WF, Bag R. Chronic Lung Allograft Dysfunction. CURRENT PULMONOLOGY REPORTS 2018. [DOI: 10.1007/s13665-018-0208-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
27
|
Young KA, Dilling DF. The Future of Lung Transplantation. Chest 2018; 155:465-473. [PMID: 30171860 DOI: 10.1016/j.chest.2018.08.1036] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
The field of lung transplant has made significant advances over the last several decades. Despite these advances, morbidity and mortality remain high when compared with other solid organ transplants. As the field moves forward, the speed by which progress can be made will in part be determined by our ability to overcome several stumbling blocks, including donor shortage, proper selection of candidates, primary graft dysfunction, and chronic lung allograft dysfunction. The advances and developments surrounding these factors will have a significant impact on shaping the field within the coming years. In this review, we look at the current climate (ripe for expanding the donor pool), new technology (ex vivo lung perfusion and bioengineered lungs), cutting-edge innovation (novel biomarkers and new ways to treat infected donors), and evidence-based medicine to discuss current trends and predict future developments for what we hope is a bright future for the field of lung transplantation.
Collapse
Affiliation(s)
- Katherine A Young
- Department of Pulmonary and Critical Care, Loyola University Medical Center, Maywood, IL
| | - Daniel F Dilling
- Department of Pulmonary and Critical Care, Loyola University Medical Center, Maywood, IL.
| |
Collapse
|
28
|
Ali HA, Pavlisko EN, Snyder LD, Frank M, Palmer SM. Complement system in lung transplantation. Clin Transplant 2018; 32:e13208. [DOI: 10.1111/ctr.13208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Hakim Azfar Ali
- Division of Pulmonary, Allergy and Critical Care; Department of Medicine; Duke University Hospital; Durham NC USA
| | | | - Laurie D. Snyder
- Division of Pulmonary, Allergy and Critical Care; Department of Medicine; Duke University Hospital; Durham NC USA
| | - Michael Frank
- Department of Pediatrics; Duke University Hospital; Durham NC USA
| | - Scott M. Palmer
- Division of Pulmonary, Allergy and Critical Care; Department of Medicine; Duke University Hospital; Durham NC USA
| |
Collapse
|
29
|
Verleden SE, Vanaudenaerde BM, Emonds MP, Van Raemdonck DE, Neyrinck AP, Verleden GM, Vos R. Donor-specific and -nonspecific HLA antibodies and outcome post lung transplantation. Eur Respir J 2017; 50:50/5/1701248. [PMID: 29146602 DOI: 10.1183/13993003.01248-2017] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/11/2017] [Indexed: 12/31/2022]
Abstract
Donor-specific antibodies (DSAs) against human leukocyte antigen (HLA) are associated with chronic lung allograft dysfunction (CLAD) and mortality post lung transplantation, but data concerning prevalence, time of onset, persistence and effects on long-term outcome remain scarce.We assessed the association between HLA antibodies and CLAD-free and graft survival in a cohort of 362 patients. We stratified our analysis according to DSA status, persistence of antibodies and timing of antibodies (pre-transplant, early or late post-transplant).Within our cohort, 61 (17%) patients developed DSAs (mostly against HLA-DQ), which was associated with worse CLAD-free and graft survival (p<0.0001 and p=0.059, respectively). Persistent (hazard ratio (HR) 3.386, 95% CI 1.928-5.948; p<0.0001) as well as transient (HR 2.998, 95% CI 1.406-6.393; p=0.0045) DSAs were associated with shorter CLAD-free survival compared with patients without DSAs. Persistent DSAs (HR 3.071, 95% CI 1.632-5.778; p=0.0005) but not transient DSAs were negatively associated with graft survival compared with patients without DSAs, likely due to the higher incidence of restrictive CLAD. HLA non-DSAs and pre-transplant HLA antibodies had no effect on post-transplant outcome.We demonstrated an important difference in prognosis between persistent and transient DSAs. Moreover, the observed association between DSAs and restrictive CLAD suggests an overlap between antibody-mediated rejection and restrictive CLAD that needs further investigation.
Collapse
Affiliation(s)
- Stijn E Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Marie-Paul Emonds
- HILA laboratory, Rode Kruis Vlaanderen, Mechelen, Belgium.,Dept of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Division of Respiratory Disease, Dept of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Verleden SE, Vos R, Vanaudenaerde BM, Verleden GM. Chronic lung allograft dysfunction phenotypes and treatment. J Thorac Dis 2017; 9:2650-2659. [PMID: 28932572 DOI: 10.21037/jtd.2017.07.81] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle limiting long-term survival post lung transplantation. Given the clinical heterogeneity of CLAD, recently two phenotypes of CLAD have been defined [bronchiolitis obliterans syndrome (BOS) vs. restrictive allograft syndrome (RAS) or restrictive CLAD (rCLAD)]. BOS is characterized by an obstructive pulmonary function, air trapping on CT and obliterative bronchiolitis (OB) on histopathology, while RAS/rCLAD patients show a restrictive pulmonary function, persistent pleuro-parenchymal infiltrates on CT and pleuroparenchymal fibro-elastosis on biopsies. Importantly, the patients with RAS/rCLAD have a severely limited survival post diagnosis of 6-18 months compared to 3-5 years after BOS diagnosis. In this review, we will review historical evidence for this heterogeneity and we will highlight the clinical, radiological, histopathological characteristics of both phenotypes, as well as their risk factors. Treatment of CLAD remains troublesome, nevertheless, we will give an overview of different treatment strategies that have been tried with some success. Adequate phenotyping remains difficult but is clearly needed for both clinical and scientific purposes.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Airway microbiota signals anabolic and catabolic remodeling in the transplanted lung. J Allergy Clin Immunol 2017; 141:718-729.e7. [PMID: 28729000 PMCID: PMC5792246 DOI: 10.1016/j.jaci.2017.06.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/10/2017] [Accepted: 06/13/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Homeostatic turnover of the extracellular matrix conditions the structure and function of the healthy lung. In lung transplantation, long-term management remains limited by chronic lung allograft dysfunction, an umbrella term used for a heterogeneous entity ultimately associated with pathological airway and/or parenchyma remodeling. OBJECTIVE This study assessed whether the local cross-talk between the pulmonary microbiota and host cells is a key determinant in the control of lower airway remodeling posttransplantation. METHODS Microbiota DNA and host total RNA were isolated from 189 bronchoalveolar lavages obtained from 116 patients post lung transplantation. Expression of a set of 11 genes encoding either matrix components or factors involved in matrix synthesis or degradation (anabolic and catabolic remodeling, respectively) was quantified by real-time quantitative PCR. Microbiota composition was characterized using 16S ribosomal RNA gene sequencing and culture. RESULTS We identified 4 host gene expression profiles, among which catabolic remodeling, associated with high expression of metallopeptidase-7, -9, and -12, diverged from anabolic remodeling linked to maximal thrombospondin and platelet-derived growth factor D expression. While catabolic remodeling aligned with a microbiota dominated by proinflammatory bacteria (eg, Staphylococcus, Pseudomonas, and Corynebacterium), anabolic remodeling was linked to typical members of the healthy steady state (eg, Prevotella, Streptococcus, and Veillonella). Mechanistic assays provided direct evidence that these bacteria can impact host macrophage-fibroblast activation and matrix deposition. CONCLUSIONS Host-microbes interplay potentially determines remodeling activities in the transplanted lung, highlighting new therapeutic opportunities to ultimately improve long-term lung transplant outcome.
Collapse
|
32
|
Vandermeulen E, Lammertyn E, Verleden SE, Ruttens D, Bellon H, Ricciardi M, Somers J, Bracke KR, Van Den Eynde K, Tousseyn T, Brusselle GG, Verbeken EK, Verschakelen J, Emonds MP, Van Raemdonck DE, Verleden GM, Vos R, Vanaudenaerde BM. Immunological diversity in phenotypes of chronic lung allograft dysfunction: a comprehensive immunohistochemical analysis. Transpl Int 2016; 30:134-143. [PMID: 27933655 DOI: 10.1111/tri.12882] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 11/30/2022]
Abstract
Chronic rejection after organ transplantation is defined as a humoral- and cell-mediated immune response directed against the allograft. In lung transplantation, chronic rejection is nowadays clinically defined as a cause of chronic lung allograft dysfunction (CLAD), consisting of different clinical phenotypes including restrictive allograft syndrome (RAS) and bronchiolitis obliterans syndrome (BOS). However, the differential role of humoral and cellular immunity is not investigated up to now. Explant lungs of patients with end-stage BOS (n = 19) and RAS (n = 18) were assessed for the presence of lymphoid (B and T cells) and myeloid cells (dendritic cells, eosinophils, mast cells, neutrophils, and macrophages) and compared to nontransplant control lung biopsies (n = 21). All myeloid cells, with exception of dendritic cells, were increased in RAS versus control (neutrophils, eosinophils, and mast cells: all P < 0.05, macrophages: P < 0.001). Regarding lymphoid cells, B cells and cytotoxic T cells were increased remarkably in RAS versus control (P < 0.001) and in BOS versus control (P < 0.01). Interestingly, lymphoid follicles were restricted to RAS (P < 0.001 versus control and P < 0.05 versus BOS). Our data suggest an immunological diversity between BOS and RAS, with a more pronounced involvement of the B-cell response in RAS characterized by a structural organization of lymphoid follicles. This may impact future therapeutic approaches.
Collapse
Affiliation(s)
- Elly Vandermeulen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Elise Lammertyn
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Stijn E Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - David Ruttens
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Hannelore Bellon
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Mario Ricciardi
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Jana Somers
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Ken R Bracke
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathleen Van Den Eynde
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Thomas Tousseyn
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Erik K Verbeken
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Johny Verschakelen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | | | - Dirk E Van Raemdonck
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| |
Collapse
|