1
|
Mohanty RP, Moghbeli K, Singer JP, Calabrese DR, Hays SR, Iasella C, Lieber S, Leard LE, Shah RJ, Venado A, Kleinhenz ME, Golden JA, Martinu T, Love C, Ward R, Langelier CR, McDyer J, Greenland JR. Small airway brush gene expression predicts chronic lung allograft dysfunction and mortality. J Heart Lung Transplant 2024; 43:1820-1832. [PMID: 39115489 DOI: 10.1016/j.healun.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) limits survival following lung transplant, but substantial lung damage occurs before diagnosis by traditional methods. We hypothesized that small airway gene expression patterns could identify CLAD risk before spirometric diagnosis and predict subsequent graft failure. METHODS Candidate genes from 4 rejection-associated transcript sets were assessed for associations with CLAD or graft failure in a derivation cohort of 156 small airway brushes from 45 CLAD cases and 37 time-matched controls with >1-year stable lung function. Candidate genes not associated with CLAD and time to graft failure were excluded, yielding the Airway Inflammation 2 (AI2) gene set. Area under the receiver operating curve (AUC) for CLAD and competing risks of death or graft failure were assessed in an independent validation cohort of 37 CLAD cases and 37 controls. RESULTS Thirty-two candidate genes were associated with CLAD and graft failure, comprising the AI2 score, which clustered into 3 subcomponents. The AI2 score identified CLAD before its onset, in early and late post-CLAD brushes, as well as in the validation cohort (AUC 0.69-0.88). The AI2 score association with CLAD was independent of positive microbiology, CLAD stage, or CLAD subtype. However, transcripts most associated with CLAD evolved over time from CLAD onset. The AI2 score predicted time to graft failure and retransplant-free survival in both cohorts (p ≤ 0.03). CONCLUSIONS This airway inflammation gene score is associated with CLAD development, graft failure, and death. Future studies defining the molecular heterogeneity of airway inflammation could lead to endotype-targeted therapies.
Collapse
Affiliation(s)
- Rashmi Prava Mohanty
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Kaveh Moghbeli
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Steven R Hays
- Department of Medicine, University of California, San Francisco, California
| | - Carlo Iasella
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sophia Lieber
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lorriana E Leard
- Department of Medicine, University of California, San Francisco, California
| | - Rupal J Shah
- Department of Medicine, University of California, San Francisco, California
| | - Aida Venado
- Department of Medicine, University of California, San Francisco, California
| | - Mary E Kleinhenz
- Department of Medicine, University of California, San Francisco, California
| | - Jeffery A Golden
- Department of Medicine, University of California, San Francisco, California
| | - Tereza Martinu
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Christina Love
- Department of Medicine, University of California, San Francisco, California
| | - Ryan Ward
- Department of Medicine, University of California, San Francisco, California
| | | | - John McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California.
| |
Collapse
|
2
|
Martin C, Mahan KS, Wiggen TD, Gilbertsen AJ, Hertz MI, Hunter RC, Quinn RA. Microbiome and metabolome patterns after lung transplantation reflect underlying disease and chronic lung allograft dysfunction. MICROBIOME 2024; 12:196. [PMID: 39385282 PMCID: PMC11462767 DOI: 10.1186/s40168-024-01893-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Progression of chronic lung disease may lead to the requirement for lung transplant (LTx). Despite improvements in short-term survival after LTx, chronic lung allograft dysfunction (CLAD) remains a critical challenge for long-term survival. This study investigates the molecular and microbial relationships between underlying lung disease and the development of CLAD in bronchoalveolar lavage fluid (BALF) from subjects post-LTx, which is crucial for tailoring treatment strategies specific to allograft dysfunctions. METHODS Paired 16S rRNA gene amplicon sequencing and untargeted LC-MS/MS metabolomics were performed on 856 BALF samples collected over 10 years from LTx recipients (n = 195) with alpha-1-antitrypsin disease (AATD, n = 23), cystic fibrosis (CF, n = 47), chronic obstructive pulmonary disease (COPD, n = 78), or pulmonary fibrosis (PF, n = 47). Data were analyzed using random forest (RF) machine learning and multivariate statistics for associations with underlying disease and CLAD development. RESULTS The BALF microbiome and metabolome after LTx differed significantly according to the underlying disease state (PERMANOVA, p = 0.001), with CF and AATD demonstrating distinct microbiome and metabolome profiles, respectively. Uniqueness in CF was mainly driven by Pseudomonas abundance and its metabolites, whereas AATD had elevated levels of phenylalanine and a lack of shared metabolites with the other underlying diseases. BALF microbiome and metabolome composition were also distinct between those who did or did not develop CLAD during the sample collection period (PERMANOVA, p = 0.001). An increase in the average abundance of Veillonella (AATD, COPD) and Streptococcus (CF, PF) was associated with CLAD development, and decreases in the abundance of phenylalanine-derivative alkaloids (CF, COPD) and glycerophosphorylcholines (CF, COPD, PF) were signatures of the CLAD metabolome. Although the relative abundance of Pseudomonas was not associated with CLAD, the abundance of its virulence metabolites, including siderophores, quorum-sensing quinolones, and phenazines, were elevated in those with CF who developed CLAD. There was a positive correlation between the abundance of these molecules and the abundance of Pseudomonas in the microbiome, but there was no correlation between their abundance and the time in which BALF samples were collected post-LTx. CONCLUSIONS The BALF microbiome and metabolome after LTx are particularly distinct in those with underlying CF and AATD. These data reflect those who developed CLAD, with increased virulence metabolite production from Pseudomonas, an aspect of CF CLAD cases. These findings shed light on disease-specific microbial and metabolic signatures in LTx recipients, offering valuable insights into the underlying causes of allograft rejection. Video Abstract.
Collapse
Affiliation(s)
- Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Kathleen S Mahan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Talia D Wiggen
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Adam J Gilbertsen
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Marshall I Hertz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Ryan C Hunter
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14051, USA.
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
3
|
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
|
4
|
Novo M, Nordén R, Westin J, Dellgren G, Böhmer J, Ricksten A, Magnusson JM. Donor Fractions of Cell-Free DNA Are Elevated During CLAD But Not During Infectious Complications After Lung Transplantation. Transpl Int 2024; 37:12772. [PMID: 39114640 PMCID: PMC11303165 DOI: 10.3389/ti.2024.12772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
During the last few years, cell-free DNA (cfDNA) has emerged as a possible non-invasive biomarker for prediction of complications after lung transplantation. We previously published a proof-of-concept study using a digital droplet polymerase chain reaction (ddPCR)-based method for detection of cfDNA. In the current study, we aimed to further evaluate the potential clinical usefulness of detecting chronic lung allograft dysfunction (CLAD) using three different ddPCR applications measuring and calculating the donor fraction (DF) of cfDNA as well as one method using the absolute amount of donor-derived cfDNA. We analyzed 246 serum samples collected from 26 lung transplant recipients. Nine of the patients had ongoing CLAD at some point during follow-up. All four methods showed statistically significant elevation of the measured variable in the CLAD samples compared to the non-CLAD samples. The results support the use of ddPCR-detected cfDNA as a potential biomarker for prediction of CLAD. These findings need to be validated in a subsequent prospective study.
Collapse
Affiliation(s)
- Mirza Novo
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rickard Nordén
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Westin
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Göran Dellgren
- Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jens Böhmer
- Pediatric Heart Center, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Clinic Frankfurt-Höchst, Frankfurt, Germany
| | - Anne Ricksten
- Department of Clinical Genetics and Genomics, Sahlgrenska Academy, University of Gothenbururg, Gothenburg, Sweden
| | - Jesper M. Magnusson
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
5
|
Figueiredo C, Chen-Wacker C, Salman J, Carvalho-Oliveira M, Monthé TS, Höffler K, Rother T, Hacker K, Valdivia E, Pogozhykh O, Hammer S, Sommer W, Yuzefovych Y, Wenzel N, Haverich A, Warnecke G, Blasczyk R. Knockdown of swine leukocyte antigen expression in porcine lung transplants enables graft survival without immunosuppression. Sci Transl Med 2024; 16:eadi9548. [PMID: 39018368 DOI: 10.1126/scitranslmed.adi9548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 12/21/2023] [Accepted: 06/21/2024] [Indexed: 07/19/2024]
Abstract
Immune rejection remains the major obstacle to long-term survival of allogeneic lung transplants. The expression of major histocompatibility complex molecules and minor histocompatibility antigens triggers allogeneic immune responses that can lead to allograft rejection. Transplant outcomes therefore depend on long-term immunosuppression, which is associated with severe side effects. To address this problem, we investigated the effect of genetically engineered transplants with permanently down-regulated swine leukocyte antigen (SLA) expression to prevent rejection in a porcine allogeneic lung transplantation (LTx) model. Minipig donor lungs with unmodified SLA expression (control group, n = 7) or with modified SLA expression (treatment group, n = 7) were used to evaluate the effects of SLA knockdown on allograft survival and on the nature and strength of immune responses after terminating an initial 4-week period of immunosuppression after LTx. Genetic engineering to down-regulate SLA expression was achieved during ex vivo lung perfusion by lentiviral transduction of short hairpin RNAs targeting mRNAs encoding β2-microglobulin and class II transactivator. Whereas all grafts in the control group were rejected within 3 months, five of seven animals in the treatment group maintained graft survival without immunosuppression during the 2-year monitoring period. Compared with controls, SLA-silenced lung recipients had lower donor-specific antibodies and proinflammatory cytokine concentrations in the serum. Together, these data demonstrate a survival benefit of SLA-down-regulated lung transplants in the absence of immunosuppression.
Collapse
Affiliation(s)
- Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Chen Chen-Wacker
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Jawad Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Marco Carvalho-Oliveira
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | | | - Klaus Höffler
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Tamina Rother
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Karolin Hacker
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Emilio Valdivia
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Olena Pogozhykh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Hammer
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Wiebke Sommer
- Department of Cardiac Surgery, University of Kiel, 24105 Kiel, Germany
| | - Yuliia Yuzefovych
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Nadine Wenzel
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Gregor Warnecke
- Department of Cardiac Surgery, University of Kiel, 24105 Kiel, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
6
|
Bogyó LZ, Török K, Illés Z, Szilvási A, Székely B, Bohács A, Pipek O, Madurka I, Megyesfalvi Z, Rényi-Vámos F, Döme B, Bogos K, Gieszer B, Bakos E. Pseudomonas aeruginosa infection correlates with high MFI donor-specific antibody development following lung transplantation with consequential graft loss and shortened CLAD-free survival. Respir Res 2024; 25:262. [PMID: 38951782 PMCID: PMC11218249 DOI: 10.1186/s12931-024-02868-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Donor-specific antibodies (DSAs) are common following lung transplantation (LuTx), yet their role in graft damage is inconclusive. Mean fluorescent intensity (MFI) is the main read-out of DSA diagnostics; however its value is often disregarded when analyzing unwanted post-transplant outcomes such as graft loss or chronic lung allograft dysfunction (CLAD). Here we aim to evaluate an MFI stratification method in these outcomes. METHODS A cohort of 87 LuTx recipients has been analyzed, in which a cutoff of 8000 MFI has been determined for high MFI based on clinically relevant data. Accordingly, recipients were divided into DSA-negative, DSA-low and DSA-high subgroups. Both graft survival and CLAD-free survival were evaluated. Among factors that may contribute to DSA development we analyzed Pseudomonas aeruginosa (P. aeruginosa) infection in bronchoalveolar lavage (BAL) specimens. RESULTS High MFI DSAs contributed to clinical antibody-mediated rejection (AMR) and were associated with significantly worse graft (HR: 5.77, p < 0.0001) and CLAD-free survival (HR: 6.47, p = 0.019) compared to low or negative MFI DSA levels. Analysis of BAL specimens revealed a strong correlation between DSA status, P. aeruginosa infection and BAL neutrophilia. DSA-high status and clinical AMR were both independent prognosticators for decreased graft and CLAD-free survival in our multivariate Cox-regression models, whereas BAL neutrophilia was associated with worse graft survival. CONCLUSIONS P. aeruginosa infection rates are elevated in recipients with a strong DSA response. Our results indicate that the simultaneous interpretation of MFI values and BAL neutrophilia is a feasible approach for risk evaluation and may help clinicians when to initiate DSA desensitization therapy, as early intervention could improve prognosis.
Collapse
Affiliation(s)
- Levente Zoltán Bogyó
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
| | - Klára Török
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
| | - Zsuzsanna Illés
- Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Anikó Szilvási
- Hungarian National Blood Transfusion Service, Budapest, Hungary
| | - Bálint Székely
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
| | - Anikó Bohács
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Orsolya Pipek
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- Department of Physics of Complex Systems, Eotvos Loránd University, Budapest, Hungary
| | - Ildikó Madurka
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Ferenc Rényi-Vámos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
- National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
| | - Balázs Döme
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Krisztina Bogos
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary.
| | - Balázs Gieszer
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary.
- National Korányi Institute of Pulmonology, Koranyi Frigyes ut 1, Budapest, 1121, Hungary.
| | - Eszter Bakos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
| |
Collapse
|
7
|
Calabrese DR, Ekstrand CA, Yellamilli S, Singer JP, Hays SR, Leard LE, Shah RJ, Venado A, Kolaitis NA, Perez A, Combes A, Greenland JR. Macrophage and CD8 T cell discordance are associated with acute lung allograft dysfunction progression. J Heart Lung Transplant 2024; 43:1074-1086. [PMID: 38367738 PMCID: PMC11230518 DOI: 10.1016/j.healun.2024.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Acute lung allograft dysfunction (ALAD) is an imprecise syndrome denoting concern for the onset of chronic lung allograft dysfunction (CLAD). Mechanistic biomarkers are needed that stratify risk of ALAD progression to CLAD. We hypothesized that single cell investigation of bronchoalveolar lavage (BAL) cells at the time of ALAD would identify immune cells linked to progressive graft dysfunction. METHODS We prospectively collected BAL from consenting lung transplant recipients for single cell RNA sequencing. ALAD was defined by a ≥10% decrease in FEV1 not caused by infection or acute rejection and samples were matched to BAL from recipients with stable lung function. We examined cell compositional and transcriptional differences across control, ALAD with decline, and ALAD with recovery groups. We also assessed cell-cell communication. RESULTS BAL was assessed for 17 ALAD cases with subsequent decline (ALAD declined), 13 ALAD cases that resolved (ALAD recovered), and 15 cases with stable lung function. We observed broad differences in frequencies of the 26 unique cell populations across groups (p = 0.02). A CD8 T cell (p = 0.04) and a macrophage cluster (p = 0.01) best identified ALAD declined from the ALAD recovered and stable groups. This macrophage cluster was distinguished by an anti-inflammatory signature and the CD8 T cell cluster resembled a Tissue Resident Memory subset. Anti-inflammatory macrophages signaled to activated CD8 T cells via class I HLA, fibronectin, and galectin pathways (p < 0.05 for each). Recipients with discordance between these cells had a nearly 5-fold increased risk of severe graft dysfunction or death (HR 4.6, 95% CI 1.1-19.2, adjusted p = 0.03). We validated these key findings in 2 public lung transplant genomic datasets. CONCLUSIONS BAL anti-inflammatory macrophages may protect against CLAD by suppressing CD8 T cells. These populations merit functional and longitudinal assessment in additional cohorts.
Collapse
Affiliation(s)
- Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California.
| | | | - Shivaram Yellamilli
- Department of Pathology, University of California, San Francisco, California
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R Hays
- Department of Medicine, University of California, San Francisco, California
| | - Lorriana E Leard
- Department of Medicine, University of California, San Francisco, California
| | - Rupal J Shah
- Department of Medicine, University of California, San Francisco, California
| | - Aida Venado
- Department of Medicine, University of California, San Francisco, California
| | | | - Alyssa Perez
- Department of Medicine, University of California, San Francisco, California
| | - Alexis Combes
- Department of Pathology, University of California, San Francisco, California
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
8
|
Kaes J, Pollenus E, Hooft C, Liu H, Aelbrecht C, Cambier S, Jin X, Van Slambrouck J, Beeckmans H, Kerckhof P, Velde GV, Van Raemdonck D, Yildirim AÖ, Van den Steen PE, Vos R, Ceulemans LJ, Vanaudenaerde BM. The Immunopathology of Pulmonary Rejection after Murine Lung Transplantation. Cells 2024; 13:241. [PMID: 38334633 PMCID: PMC10854916 DOI: 10.3390/cells13030241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
To improve outcomes following lung transplantation, it is essential to understand the immunological mechanisms that result in chronic graft failure. The associated clinical syndrome is termed chronic lung allograft dysfunction (CLAD), which is known to be induced by alloimmune-dependent (i.e., rejection) and alloimmune-independent factors (e.g., infections, reflux and environmental factors). We aimed to explore the alloimmune-related mechanism, i.e., pulmonary rejection. In this study, we use a murine orthotopic left lung transplant model using isografts and allografts (C57BL/6 or BALB/c as donors to C57BL/6 recipients), with daily immunosuppression (10 mg/kg cyclosporin A and 1.6 mg/kg methylprednisolone). Serial sacrifice was performed at days 1, 7 and 35 post-transplantation (n = 6 at each time point for each group). Left transplanted lungs were harvested, a single-cell suspension was made and absolute numbers of immune cells were quantified using multicolor flow cytometry. The rejection process followed the principles of a classic immune response, including innate but mainly adaptive immune cells. At day 7 following transplantation, the numbers of interstitial macrophages, monocytes, dendritic cells, NK cells, NKT cells, CD4+ T cells and CD8+ T and B cells were increased in allografts compared with isografts. Only dendritic cells and CD4+ T cells remained elevated at day 35 in allografts. Our study provides insights into the immunological mechanisms of true pulmonary rejection after murine lung transplantation. These results might be important in further research on diagnostic evaluation and treatment for CLAD.
Collapse
Affiliation(s)
- Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium; (E.P.)
| | - Charlotte Hooft
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Hengshuo Liu
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany (A.Ö.Y.)
| | - Celine Aelbrecht
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium;
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany (A.Ö.Y.)
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium; (E.P.)
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.K.)
| |
Collapse
|
9
|
Bansal S, Fleming T, Canez J, Maine GN, Bharat A, Walia R, Tokman S, Smith MA, Tiffany B, Bremner RM, Mohanakumar T. Immune responses of lung transplant recipients against SARS-CoV-2 and common respiratory coronaviruses: Evidence for pre-existing cross-reactive immunity. Transpl Immunol 2023; 81:101940. [PMID: 37866672 PMCID: PMC11019873 DOI: 10.1016/j.trim.2023.101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Humoral and cellular immune responses to SARS-CoV-2 and other coronaviruses in lung transplant recipients are unknown. We measured antibodies and T cell responses against the SARS-CoV-2 spike S2 and nucleocapsid antigens and spike antigens from common respiratory coronaviruses (229E, NL63, OC43, and HKU1) after vaccination or infection of LTxRs. 148 LTxRs from single center were included in this study: 98 after vaccination and 50 following SARS-CoV-2 infection. Antibodies were quantified by enzyme-linked immunosorbent assay. The frequency of T cells secreting IL2, IL4, IL10, IL17, TNFα, and IFNγ were enumerated by enzyme-linked immunospot assay. Our results have shown the development of antibodies to SARS-CoV-2 spike protein in infected LTxRs (39/50) and vaccinated LTxRs (52/98). Vaccinated LTxRs had higher number of T cells producing TNFα but less cells producing IFNγ than infected LTxRs in response to the nucleocapsid antigen and other coronavirus spike antigens. We didn't find correlation between the development of antibodies and cellular immune responses against the SARS-CoV-2 spike protein after vaccination. Instead, LTxRs have pre-existing cellular immunity to common respiratory coronaviruses, leading to cross-reactive immunity against SARS-CoV-2 which likely will provide protection against SARS-Cov-2 infection.
Collapse
Affiliation(s)
- Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Jesse Canez
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Gabriel N Maine
- Department of Pathology and Laboratory Medicine, Royal Oak, Beaumont Health, MI, USA
| | | | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Brian Tiffany
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - T Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| |
Collapse
|
10
|
Li DJ, Abele J, Sunner P, Varughese RA, Hirji AS, Weinkauf JG, Nagendran J, Weatherald JC, Lien DC, Halloran KM. Relative Lung Perfusion on Ventilation-Perfusion Scans After Double Lung Transplant. Transplantation 2023; 107:2262-2270. [PMID: 37291709 DOI: 10.1097/tp.0000000000004683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND Pulmonary blood flow can be assessed on ventilation-perfusion (VQ) scan with relative lung perfusion, with a 55% to 45% (or 10%) right-to-left differential considered normal. We hypothesized that wide perfusion differential on routine VQ studies at 3 mo posttransplant would be associated with an increased risk of death or retransplantation, chronic lung allograft (CLAD), and baseline lung allograft dysfunction. METHODS We conducted a retrospective cohort study on all patients who underwent double-lung transplant in our program between 2005 and 2016, identifying patients with a wide perfusion differential of >10% on a 3-mo VQ scan. We used Kaplan-Meier estimates and proportional hazards models to assess the association between perfusion differential and time to death or retransplant and time to CLAD onset. We used correlation and linear regression to assess the relationship with lung function at time of scan and with baseline lung allograft dysfunction. RESULTS Of 340 patients who met inclusion criteria, 169 (49%) had a relative perfusion differential of ≥ 10% on a 3-mo VQ scan. Patients with increased perfusion differential had increased risk of death or retransplantation ( P = 0.011) and CLAD onset ( P = 0.012) after adjustment for other radiographic/endoscopic abnormalities. Increased perfusion differential was associated with lower lung function at time of scan. CONCLUSIONS Wide lung perfusion differential was common after lung transplant in our cohort and associated with increased risk of death, poor lung function, and CLAD onset. The nature of this abnormality and its use as a predictor of future risk warrant further investigation.
Collapse
Affiliation(s)
- David J Li
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Jonathan Abele
- Department of Diagnostic Imaging and Radiology, University of Alberta, Edmonton, Canada
| | - Parveen Sunner
- Department of Diagnostic Imaging and Radiology, University of Alberta, Edmonton, Canada
| | | | - Alim S Hirji
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - Jayan Nagendran
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | - Dale C Lien
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
11
|
Halitim P, Tissot A. [Chronic lung allograft dysfunction in 2022, past and updates]. Rev Mal Respir 2023; 40:324-334. [PMID: 36858879 DOI: 10.1016/j.rmr.2023.01.025] [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: 09/21/2022] [Accepted: 01/24/2023] [Indexed: 03/03/2023]
Abstract
INTRODUCTION While short-term results of lung transplantation have improved considerably, long-term survival remains below that achieved for other solid organ transplants. CURRENT KNOWLEDGE The main cause of late mortality is chronic lung allograft dysfunction (CLAD), which affects nearly half of the recipients 5 years after transplantation. Immunological and non-immune risk factors have been identified. These factors activate the innate and adaptive immune system, leading to lesional and altered wound-healing processes, which result in fibrosis affecting the small airways or interstitial tissue. Several phenotypes of CLAD have been identified based on respiratory function and imaging pattern. Aside from retransplantation, which is possible for only small number of patients, no treatment can reverse the CLAD process. PERSPECTIVES Current therapeutic research is focused on anti-fibrotic treatments and photopheresis. Basic research has identified numerous biomarkers that could prove to be relevant as therapeutic targets. CONCLUSION While the pathophysiological mechanisms of CLAD are better understood than before, a major therapeutic challenge remains.
Collapse
Affiliation(s)
- P Halitim
- Service de pneumologie et soins intensifs, Hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 75015 Paris, France; Service de pneumologie, CHU de Nantes, l'Institut du thorax, Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44093 Nantes cedex, France
| | - A Tissot
- Service de pneumologie, CHU de Nantes, l'Institut du thorax, Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44093 Nantes cedex, France.
| |
Collapse
|
12
|
D'Angelo D, Quarta E, Glieca S, Varacca G, Flammini L, Bertoni S, Brandolini M, Sambri V, Grumiro L, Gatti G, Dirani G, Taddei F, Bianchera A, Sonvico F, Bettini R, Buttini F. An Enhanced Dissolving Cyclosporin-A Inhalable Powder Efficiently Reduces SARS-CoV-2 Infection In Vitro. Pharmaceutics 2023; 15:pharmaceutics15031023. [PMID: 36986883 PMCID: PMC10055879 DOI: 10.3390/pharmaceutics15031023] [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: 02/08/2023] [Revised: 03/08/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
This work illustrates the development of a dry inhalation powder of cyclosporine-A for the prevention of rejection after lung transplantation and for the treatment of COVID-19. The influence of excipients on the spray-dried powder's critical quality attributes was explored. The best-performing powder in terms of dissolution time and respirability was obtained starting from a concentration of ethanol of 45% (v/v) in the feedstock solution and 20% (w/w) of mannitol. This powder showed a faster dissolution profile (Weibull dissolution time of 59.5 min) than the poorly soluble raw material (169.0 min). The powder exhibited a fine particle fraction of 66.5% and an MMAD of 2.97 µm. The inhalable powder, when tested on A549 and THP-1, did not show cytotoxic effects up to a concentration of 10 µg/mL. Furthermore, the CsA inhalation powder showed efficiency in reducing IL-6 when tested on A549/THP-1 co-culture. A reduction in the replication of SARS-CoV-2 on Vero E6 cells was observed when the CsA powder was tested adopting the post-infection or simultaneous treatment. This formulation could represent a therapeutic strategy for the prevention of lung rejection, but is also a viable approach for the inhibition of SARS-CoV-2 replication and the COVID-19 pulmonary inflammatory process.
Collapse
Affiliation(s)
- Davide D'Angelo
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Eride Quarta
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Stefania Glieca
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Giada Varacca
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Lisa Flammini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Simona Bertoni
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Martina Brandolini
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, 40138 Bologna, Italy
- Microbiology Unit, The Great Romagna Area Hub Laboratory, Piazza della Liberazione 60, Pievesestina, 47522 Cesena, Italy
| | - Vittorio Sambri
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, 40138 Bologna, Italy
- Microbiology Unit, The Great Romagna Area Hub Laboratory, Piazza della Liberazione 60, Pievesestina, 47522 Cesena, Italy
| | - Laura Grumiro
- Microbiology Unit, The Great Romagna Area Hub Laboratory, Piazza della Liberazione 60, Pievesestina, 47522 Cesena, Italy
| | - Giulia Gatti
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, 40138 Bologna, Italy
| | - Giorgio Dirani
- Microbiology Unit, The Great Romagna Area Hub Laboratory, Piazza della Liberazione 60, Pievesestina, 47522 Cesena, Italy
| | - Francesca Taddei
- Microbiology Unit, The Great Romagna Area Hub Laboratory, Piazza della Liberazione 60, Pievesestina, 47522 Cesena, Italy
| | - Annalisa Bianchera
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Fabio Sonvico
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Ruggero Bettini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27a, 43124 Parma, Italy
| |
Collapse
|
13
|
Pison C, Tissot A, Bernasconi E, Royer PJ, Roux A, Koutsokera A, Coiffard B, Renaud-Picard B, Le Pavec J, Mordant P, Demant X, Villeneuve T, Mornex JF, Nemska S, Frossard N, Brugière O, Siroux V, Marsland BJ, Foureau A, Botturi K, Durand E, Pellet J, Danger R, Auffray C, Brouard S, Nicod L, Magnan A. Systems prediction of chronic lung allograft dysfunction: Results and perspectives from the Cohort of Lung Transplantation and Systems prediction of Chronic Lung Allograft Dysfunction cohorts. Front Med (Lausanne) 2023; 10:1126697. [PMID: 36968829 PMCID: PMC10033762 DOI: 10.3389/fmed.2023.1126697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
BackgroundChronic lung allograft dysfunction (CLAD) is the leading cause of poor long-term survival after lung transplantation (LT). Systems prediction of Chronic Lung Allograft Dysfunction (SysCLAD) aimed to predict CLAD.MethodsTo predict CLAD, we investigated the clinicome of patients with LT; the exposome through assessment of airway microbiota in bronchoalveolar lavage cells and air pollution studies; the immunome with works on activation of dendritic cells, the role of T cells to promote the secretion of matrix metalloproteinase-9, and subpopulations of T and B cells; genome polymorphisms; blood transcriptome; plasma proteome studies and assessment of MSK1 expression.ResultsClinicome: the best multivariate logistic regression analysis model for early-onset CLAD in 422 LT eligible patients generated a ROC curve with an area under the curve of 0.77. Exposome: chronic exposure to air pollutants appears deleterious on lung function levels in LT recipients (LTRs), might be modified by macrolides, and increases mortality. Our findings established a link between the lung microbial ecosystem, human lung function, and clinical stability post-transplant. Immunome: a decreased expression of CLEC1A in human lung transplants is predictive of the development of chronic rejection and associated with a higher level of interleukin 17A; Immune cells support airway remodeling through the production of plasma MMP-9 levels, a potential predictive biomarker of CLAD. Blood CD9-expressing B cells appear to favor the maintenance of long-term stable graft function and are a potential new predictive biomarker of BOS-free survival. An early increase of blood CD4 + CD57 + ILT2+ T cells after LT may be associated with CLAD onset. Genome: Donor Club cell secretory protein G38A polymorphism is associated with a decreased risk of severe primary graft dysfunction after LT. Transcriptome: blood POU class 2 associating factor 1, T-cell leukemia/lymphoma domain, and B cell lymphocytes, were validated as predictive biomarkers of CLAD phenotypes more than 6 months before diagnosis. Proteome: blood A2MG is an independent predictor of CLAD, and MSK1 kinase overexpression is either a marker or a potential therapeutic target in CLAD.ConclusionSystems prediction of Chronic Lung Allograft Dysfunction generated multiple fingerprints that enabled the development of predictors of CLAD. These results open the way to the integration of these fingerprints into a predictive handprint.
Collapse
Affiliation(s)
- Christophe Pison
- Service Hospitalier Universitaire de Pneumologie Physiologie, Pôle Thorax et Vaisseaux, Fédération Grenoble Transplantation, CHU Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, INSERM 1055, Grenoble, France
- *Correspondence: Christophe Pison,
| | - Adrien Tissot
- Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Eric Bernasconi
- Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse
| | - Pierre-Joseph Royer
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Antoine Roux
- Service de Pneumologie, Hôpital Foch, Suresnes, France
- Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement, INRAE, Jouy-en-Josas, France
| | - Angela Koutsokera
- Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse
| | - Benjamin Coiffard
- Service de Pneumologie et de Transplantation Pulmonaire, APHM, Hôpital Nord, Aix Marseille Univ, Marseille, France
| | - Benjamin Renaud-Picard
- Service de Pneumologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Inserm UMR 1260, Regenerative Nanomedicine, Université de Strasbourg, Strasbourg, France
| | - Jérôme Le Pavec
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardiopulmonaire, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Pierre Mordant
- Service de Chirurgie Vasculaire, Thoracique et Transplantation Pulmonaire, Hôpital Bichat, AP-HP, INSERM U1152, Université Paris Cité, Paris, France
| | - Xavier Demant
- Service de Pneumologie et Transplantation Pulmonaire, CHU de Bordeaux, Bordeaux, France
| | - Thomas Villeneuve
- Service de Pneumologie, CHU de Toulouse, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Jean-Francois Mornex
- Université de Lyon, Université Lyon 1, PSL, EPHE, INRAE, IVPC, Lyon, France
- Hospices Civils de Lyon, GHE, Service de Pneumologie, RESPIFIL, Orphalung, Inserm CIC, Lyon, France
| | - Simona Nemska
- UMR 7200 - Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, CNRS-Université de Strasbourg, Illkirch, France
| | - Nelly Frossard
- UMR 7200 - Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, CNRS-Université de Strasbourg, Illkirch, France
| | - Olivier Brugière
- Service de Pneumologie, Hôpital Foch, Suresnes, France
- Laboratoire d’Immunologie de la Transplantation, Hôpital Saint-Louis, CEA/DRF/Institut de Biologie François Jacob, Unité INSERM 1152, Université Paris Diderot, USPC, Paris, France
| | - Valérie Siroux
- Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences (IAB), Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Benjamin J. Marsland
- Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Aurore Foureau
- Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Karine Botturi
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Eugenie Durand
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Johann Pellet
- European Institute for Systems Biology and Medicine, Vourles, France
| | - Richard Danger
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, Vourles, France
| | - Sophie Brouard
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Laurent Nicod
- Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse
| | - Antoine Magnan
- Service de Pneumologie, Hôpital Foch, Suresnes, France
- Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement, INRAE, Jouy-en-Josas, France
| | | |
Collapse
|
14
|
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
|
15
|
Ju C, Xu X, Zhang J, Chen A, Lian Q, Liu F, Liu H, Cai Y, Zou Y, Yang Y, Zhou Y, He J. Application of plasma donor-derived cell free DNA for lung allograft rejection diagnosis in lung transplant recipients. BMC Pulm Med 2023; 23:37. [PMID: 36703125 PMCID: PMC9881379 DOI: 10.1186/s12890-022-02229-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/07/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Donor-derived cell-free DNA (dd-cfDNA) has been applied to monitor acute rejection (AR) in kidney and heart transplantation. This study was aimed to investigate the application of dd-cfDNA levels in the diagnosis of AR and chronic lung allograft dysfunction (CLAD) among the lung transplantation recipients (LTRs). METHODS One hundred and seventy LTRs were enrolled at the First Affiliated Hospital of Guangzhou Medical University between 1 June 2015 and 30 March 2021. Patients were divided into 4 groups: stable group, AR group, infection group and CLAD group. The level of dd-cfDNA was analyzed using target region sequencing and the performance characteristics of dd-cfDNA for diagnosis of AR and CLAD were determined, respectively. RESULTS Kruskal-Wallis test showed that there were some significant differences in the level of dd-cfDNA (%) among the 4 groups, with p < 0.001. Among them, the level of dd-cfDNA (%) was highest (median 2.17, IQR [1.40-3.82]) in AR group, and higher in CLAD group (median 1.07, IQR [0.98-1.31]), but lower in infection group (median 0.71, IQR [0.57-1.07]) and lowest in stable group (median 0.71, IQR [0.61-0.84]). AUC-ROC curve analysis showed that the threshold of dd-cfDNA for AR was 1.17%, with sensitivity being 89.19% and specificity being 86.47%, and the optimal threshold of 0.89% was determined of CLAD, with sensitivity being 95.00% and specificity of 76.99%. CONCLUSIONS Plasma dd-cfDNA could be a useful tool for the assessment of lung allograft rejection, including AR and CLAD, and holds promise as a noninvasive biomarker for "allograft injury" in both acute and chronic rejection following lung transplantation.
Collapse
Affiliation(s)
- Chunrong Ju
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Xin Xu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Jianheng Zhang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Ao Chen
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Qiaoyan Lian
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Feng Liu
- AlloDx (Shanghai) Biotech., Co., Ltd, Shanghai, 201100 China
| | - Haitao Liu
- AlloDx (Shanghai) Biotech., Co., Ltd, Shanghai, 201100 China
| | - Yuhang Cai
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Yanjun Zou
- AlloDx (Shanghai) Biotech., Co., Ltd, Shanghai, 201100 China
| | - Yalan Yang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| | - Yang Zhou
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, 212013 China
| | - Jianxing He
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000 China
| |
Collapse
|
16
|
Righi I, Vaira V, Morlacchi LC, Croci GA, Rossetti V, Blasi F, Ferrero S, Nosotti M, Rosso L, Clerici M. PD-1 expression in transbronchial biopsies of lung transplant recipients is a possible early predictor of rejection. Front Immunol 2023; 13:1024021. [PMID: 36703976 PMCID: PMC9871480 DOI: 10.3389/fimmu.2022.1024021] [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: 08/20/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Chronic lung allograft dysfunction (CLAD) is the main cause of the reduced survival of lung transplanted (LTx) patients. The possible role of immune checkpoint molecules in establishing tolerance has been scarcely investigated in the setting of lung transplantation. Methods We conducted a retrospective, observational pilot study on a consecutive series of transbronchial cryobiopsies (TCB) obtained from 24 patients during LTx follow-up focusing on PD-1, one of the most investigated immune checkpoint molecules. Results Results showed that PD-1-expressing T lymphocytes were present in all TCB with a histological diagnosis of acute rejection (AR; 9/9), but not in most (11/15) of the TCB not resulting in a diagnosis of AR (p=0.0006). Notably, the presence of PD-1-expressing T lymphocytes in TCB resulted in a 10-times higher risk of developing chronic lung allograft dysfunction (CLAD), the main cause of the reduced survival of lung transplanted patients, thus being associated with a clearly worst clinical outcome. Discussion Results of this pilot study indicate a central role of PD-1 in the development of AR and its evolution towards CLAD and suggest that the evaluation of PD-1-expressing lymphocytes in TCB could offer a prognostic advantage in monitoring the onset of AR in patients who underwent lung transplantation.
Collapse
Affiliation(s)
- Ilaria Righi
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Letizia Corinna Morlacchi
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Alberto Croci
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valeria Rossetti
- Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,Respiratory Unit and Adult Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Mario Nosotti
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplantation Unit, Department of Cardio- Thoracic - Vascular Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,*Correspondence: Lorenzo Rosso,
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy,Don C. Gnocchi Foundation, IRCCS, Milan, Italy
| |
Collapse
|
17
|
Trindade AJ, Chapin KC, Mullican A, Gray JN, Hoy H, Demarest CT, Lambright ES, McPherson KA, Norfolk SG, Robbins IM, Bacchetta M, Erasmus DB, Shaver CM. Assessment of dd-cfDNA Levels in Clinically Stable Lung Allograft Recipients Beyond the Initial 2 y Posttransplant. Transplant Direct 2022; 8:e1411. [PMID: 36406896 PMCID: PMC9671749 DOI: 10.1097/txd.0000000000001411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023] Open
Abstract
Donor-derived cell-free DNA (dd-cfDNA) is a useful biomarker for the diagnosis of acute allograft injury within the first 1 to 2 y after lung transplant, but its utility for diagnosing chronic lung allograft dysfunction (CLAD) has not yet been studied. Understanding baseline dd-cfDNA kinetics beyond the initial 2 y posttransplant is a necessary first step in determining the utility of dd-cfDNA as a CLAD biomarker. We seek to establish baseline dd-cfDNA% levels in clinically stable lung allograft recipients who are >2 y posttransplant. Methods We performed a prospective, single-center, observational study to identify plasma dd-cfDNA levels in clinically stable lung allograft recipients >2 y posttransplant. Results Fifty-one subjects were enrolled and ≥3 baseline dd-cfDNA measurements were acquired during a median of 252 d. The median baseline percent dd-cfDNA level in our cohort was 0.45% (interquartile range [IQR], 0.26-0.69). There were statistically significant differences in dd-cfDNA based on posttransplant duration (≤5 y posttransplant median 0.41% [IQR, 0.21-0.64] versus >5 y posttransplant median 0.50% [IQR, 0.33-0.76]; P < 0.02). However, the clinical significance of this small change in dd-cfDNA is uncertain because this magnitude of change is within the biologic test variation of 73%. Conclusions This study is the first to define levels of dd-cfDNA in clinically stable patients who are >2 y post-lung transplant. These findings lay the groundwork for the study of dd-cfDNA as a possible biomarker for CLAD.
Collapse
Affiliation(s)
- Anil J. Trindade
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Kaitlyn C. Chapin
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Amy Mullican
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Haley Hoy
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Caitlin T. Demarest
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Eric S. Lambright
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Katie A. McPherson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Stephanie G. Norfolk
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ivan M. Robbins
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew Bacchetta
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN
| | - David B. Erasmus
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
18
|
Liu CR, Heid CA, Hauptmann E, Ali M, Pruszynski J, Pillai A, Banga A, Wait MA, Huffman LC, Peltz M, Hackmann AE, Jessen ME, Ring WS, Murala JS. Donor substance use and lung transplantation: A single center experience. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
19
|
Peräkylä L, Nykänen A, Piilonen A, Kesävuori R, Halme M, Raivio P. Transition from BOS to RAS impairs prognosis after lung transplantation-CLAD subtype analysis by CT volumetry. PLoS One 2022; 17:e0275563. [PMID: 36223371 PMCID: PMC9555659 DOI: 10.1371/journal.pone.0275563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Background Chronic lung allograft dysfunction (CLAD), subclassified into bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS), limits survival after lung transplantation. Information concerning transition from BOS to RAS is limited. We aimed to characterize the lung volume change after BOS diagnosis by computed tomography (CT) volumetry and to determine the incidence, risk factors and clinical significance of BOS to RAS transition. Methods CT volumetry measurements were performed from 63 patients with CLAD initially classified as BOS by CT volumetry. BOS patients with lung volume remaining >85% of baseline were classified as persistent BOS, whereas BOS patients whose lung volume permanently decreased to ≤85% of baseline were classified as BOS to RAS transition. Results During follow-up (median 9.8 years) eight patients (12.7%) were classified as BOS to RAS transition, which decreased recipient (p = 0.004) and graft survival (p = 0.020) in comparison to patients with persistent BOS. Opacities on chest imaging preceded BOS to RAS transition in 88% of patients. Opacities on chest imaging at BOS diagnosis and early CLAD diagnosis after transplantation were risk factors for transition. Conclusion Based on lung volume decrease measured by CT volumetry, a small proportion of BOS patients transitioned to RAS which had an adverse effect on recipient and graft survival.
Collapse
Affiliation(s)
- Laura Peräkylä
- Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Antti Nykänen
- Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Anneli Piilonen
- Department of Radiology, Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Risto Kesävuori
- Department of Radiology, Medical Imaging Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Maija Halme
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Peter Raivio
- Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Fu A, Vasileva A, Hanafi N, Belousova N, Wu J, Rajyam SS, Ryan CM, Hantos Z, Chow CW. Characterization of chronic lung allograft dysfunction phenotypes using spectral and intrabreath oscillometry. Front Physiol 2022; 13:980942. [PMID: 36277208 PMCID: PMC9582781 DOI: 10.3389/fphys.2022.980942] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic lung allograft dysfunction (CLAD) is the major cause of death beyond 2 years after lung transplantation and develops in 50% of all patients by 5 years post-transplant. CLAD is diagnosed on the basis of a sustained drop of 20% for at least 3 months in the forced expiratory volume (FEV1), compared to the best baseline value achieved post-transplant. CLAD presents as two main phenotypes: bronchiolitis obliterans syndrome (BOS) is more common and has better prognosis than restrictive allograft syndrome (RAS). Respiratory oscillometry is a different modality of lung function testing that is highly sensitive to lung mechanics. The current study investigated whether spectral and intrabreath oscillometry can differentiate between CLAD-free, BOS- and RAS-CLAD at CLAD onset, i.e., at the time of the initial 20% drop in the FEV1. Methods: A retrospective, cross-sectional analysis of 263 double lung transplant recipients who underwent paired testing with oscillometry and spirometry at the Toronto General Pulmonary Function Laboratory from 2017 to 2022 was conducted. All pulmonary function testing and CLAD diagnostics were performed following international guidelines. Statistical analysis was conducted using multiple comparisons. Findings: The RAS (n = 6) spectral oscillometry pattern differs from CLAD-free (n = 225) by right-ward shift of reactance curve similar to idiopathic pulmonary fibrosis whereas BOS (n = 32) has a pattern similar to obstructive lung disease. Significant differences were found in most spectral and intrabreath parameters between BOS, RAS, and time-matched CLAD-free patients. Post-hoc analysis revealed these differences were primarily driven by BOS instead of RAS. While no differences were found between CLAD-free and RAS patients with regards to spectral oscillometry, the intrabreath metric of reactance at end-inspiration (XeI) was significantly different (p < 0.05). BOS and RAS were differentiated by spectral oscillometry measure R5, and intrabreath resistance at end expiration, ReE (p < 0.05 for both). Conclusion: Both spectral and intrabreath oscillometry can differentiate BOS-CLAD from CLAD-free states while intrabreath oscillometry, specifically XeI, can uniquely distinguish RAS-CLAD from CLAD-free. Spectral and intrabreath oscillometry offer complementary information regarding lung mechanics in CLAD patients to help distinguish the two phenotypes and could prove useful in prognostication.
Collapse
Affiliation(s)
- Anne Fu
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anastasiia Vasileva
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nour Hanafi
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Natalia Belousova
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Unit, University Health Network, Toronto, ON, Canada
| | - Joyce Wu
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Sarada Sriya Rajyam
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Clodagh M. Ryan
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Zoltán Hantos
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Chung-Wai Chow
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Unit, University Health Network, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
- *Correspondence: Chung-Wai Chow,
| |
Collapse
|
21
|
Integrated Immunologic Monitoring in Solid Organ Transplantation: The Road Toward Torque Teno Virus-guided Immunosuppression. Transplantation 2022; 106:1940-1951. [PMID: 35509090 PMCID: PMC9521587 DOI: 10.1097/tp.0000000000004153] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Potent immunosuppressive drugs have been introduced into clinical care for solid organ transplant recipients. It is now time to guide these drugs on an individual level to optimize their efficacy. An ideal tool simultaneously detects overimmunosuppression and underimmunosuppression, is highly standardized, and is straightforward to implement into routine. Randomized controlled interventional trials are crucial to demonstrate clinical value. To date, proposed assays have mainly focused on the prediction of rejection and were based on the assessment of few immune compartments. Recently, novel tools have been introduced based on a more integrated approach to characterize the immune function and cover a broader spectrum of the immune system. In this respect, the quantification of the plasma load of a highly prevalent and apathogenic virus that might reflect the immune function of its host has been proposed: the torque teno virus (TTV). Although TTV control is driven by T cells, other major immune compartments might contribute to the hosts' response. A standardized in-house polymerase chain reaction and a conformité européenne-certified commercially available polymerase chain reaction are available for TTV quantification. TTV load is associated with rejection and infection in solid organ transplant recipients, and cutoff values for risk stratification of such events have been proposed for lung and kidney transplantation. Test performance of TTV load does not allow for the diagnosis of rejection and infection but is able to define at-risk patients. Hitherto TTV load has not been used in interventional settings, but two interventional randomized controlled trials are currently testing the safety and efficacy of TTV-guided immunosuppression.
Collapse
|
22
|
Mauer J, Kuckhahn A, Ramsperger-Gleixner M, Ensminger SM, Distler JH, Weyand M, Heim C. Nintedanib reduces alloimmune-induced chronic airway changes in murine tracheal allografts. Transpl Immunol 2022; 73:101608. [DOI: 10.1016/j.trim.2022.101608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/12/2022] [Accepted: 04/23/2022] [Indexed: 01/01/2023]
|
23
|
Chronic Lung Allograft Dysfunction Is Associated with Increased Levels of Cell-Free Mitochondrial DNA in Bronchoalveolar Lavage Fluid of Lung Transplant Recipients. J Clin Med 2022; 11:jcm11144142. [PMID: 35887906 PMCID: PMC9322792 DOI: 10.3390/jcm11144142] [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] [Received: 06/05/2022] [Revised: 06/30/2022] [Accepted: 07/07/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic Lung Allograft Dysfunction (CLAD) is a life-threatening complication that limits the long-term survival of lung transplantation patients. Early diagnosis remains the basis of efficient management of CLAD, making the need for distinctive biomarkers critical. This explorative study aimed to investigate the predictive power of mitochondrial DNA (mtDNA) derived from bronchoalveolar lavages (BAL) to detect CLAD. The study included 106 lung transplant recipients and analyzed 286 BAL samples for cell count, cell differentiation, and inflammatory and mitochondrial biomarkers, including mtDNA. A receiver operating curve analysis of mtDNA levels was used to assess its ability to detect CLAD. The results revealed a discriminatory pro-inflammatory cytokine profile in the BAL fluid of CLAD patients. The concentration of mtDNA increased in step with each CLAD stage, reaching its highest concentration in stage 4, and correlated significantly with decreasing FEV1. The receiver operating curve analysis of mtDNA in BAL revealed a moderate prediction of CLAD when all stages were grouped together (AUROC 0.75, p-value < 0.0001). This study has found the concentration mtDNA in BAL to be a potential predictor for the early detection of CLAD and the differentiation of different CLAD stages, independent of the underlying pathology.
Collapse
|
24
|
Sorbini M, Togliatto G, Mioli F, Simonato E, Marro M, Cappuccio M, Arruga F, Caorsi C, Mansouri M, Magistroni P, Gambella A, Delsedime L, Papotti MG, Solidoro P, Albera C, Boffini M, Rinaldi M, Amoroso A, Vaisitti T, Deaglio S. Validation of a Simple, Rapid, and Cost-Effective Method for Acute Rejection Monitoring in Lung Transplant Recipients. Transpl Int 2022; 35:10546. [PMID: 35755857 PMCID: PMC9221674 DOI: 10.3389/ti.2022.10546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
Abstract
Despite advances in immunosuppression therapy, acute rejection remains the leading cause of graft dysfunction in lung transplant recipients. Donor-derived cell-free DNA is increasingly being considered as a valuable biomarker of acute rejection in several solid organ transplants. We present a technically improved molecular method based on digital PCR that targets the mismatch between the recipient and donor at the HLA-DRB1 locus. Blood samples collected sequentially post-transplantation from a cohort of lung recipients were used to obtain proof-of-principle for the validity of the assay, correlating results with transbronchial biopsies and lung capacity tests. The results revealed an increase in dd-cfDNA during the first 2 weeks after transplantation related to ischemia-reperfusion injury (6.36 ± 5.36%, p < 0.0001). In the absence of complications, donor DNA levels stabilized, while increasing again during acute rejection episodes (7.81 ± 12.7%, p < 0.0001). Respiratory tract infections were also involved in the release of dd-cfDNA (9.14 ± 15.59%, p = 0.0004), with a positive correlation with C-reactive protein levels. Overall, the dd-cfDNA percentages were inversely correlated with the lung function values measured by spirometry. These results confirm the value of dd-cfDNA determination during post-transplant follow-up to monitor acute rejection in lung recipients, achieved using a rapid and inexpensive approach based on the HLA mismatch between donor and recipient.
Collapse
Affiliation(s)
- Monica Sorbini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Fiorenza Mioli
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Erika Simonato
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Matteo Marro
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | | | - Francesca Arruga
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristiana Caorsi
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Morteza Mansouri
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Paola Magistroni
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | | | - Luisa Delsedime
- Pathology Unit, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Mauro Giulio Papotti
- Pathology Unit, Città Della Salute e Della Scienza University Hospital, Turin, Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Paolo Solidoro
- Lung Transplantation and Advanced Airways Management, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Carlo Albera
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Boffini
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Mauro Rinaldi
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, University of Turin, Turin, Italy.,Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy.,Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| |
Collapse
|
25
|
Tissot A, Burgel PR. No patient left behind! Therapeutic options for cystic fibrosis patients living with lung transplantation. J Cyst Fibros 2022; 21:735-736. [PMID: 35667974 DOI: 10.1016/j.jcf.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/29/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Adrien Tissot
- CHU Nantes, Service de Pneumologie, Institut du Thorax, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Pierre-Régis Burgel
- Université Paris Cité, Inserm U1016, Institut Cochin, Paris, France; Pulmonary Department and National Cystic Fibrosis Reference Centre, Cochin Hospital; Assistance Publique Hôpitaux de Paris, Paris, France.
| |
Collapse
|
26
|
Niroomand A, Ghaidan H, Hallgren O, Hansson L, Larsson H, Wagner D, Mackova M, Halloran K, Hyllén S, Lindstedt S. Corticotropin releasing hormone as an identifier of bronchiolitis obliterans syndrome. Sci Rep 2022; 12:8413. [PMID: 35589861 PMCID: PMC9120482 DOI: 10.1038/s41598-022-12546-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Lung transplantion (LTx) recipients have low long-term survival and a high incidence of bronchiolitis obliterans syndrome (BOS), an inflammation of the small airways in chronic rejection of a lung allograft. There is great clinical need for a minimally invasive biomarker of BOS. Here, 644 different proteins were analyzed to detect biomarkers that distinguish BOS grade 0 from grades 1–3. The plasma of 46 double lung transplant patients was analyzed for proteins using a high-component, multiplex immunoassay that enables analysis of protein biomarkers. Proximity Extension Assay (PEA) consists of antibody probe pairs which bind to targets. The resulting polymerase chain reaction (PCR) reporter sequence can be quantified by real-time PCR. Samples were collected at baseline and 1-year post transplantation. Enzyme-linked immunosorbent assay (ELISA) was used to validate the findings of the PEA analysis across both time points and microarray datasets from other lung transplantation centers demonstrated the same findings. Significant decreases in the plasma protein levels of CRH, FERC2, IL-20RA, TNFB, and IGSF3 and an increase in MMP-9 and CTSL1 were seen in patients who developed BOS compared to those who did not. In this study, CRH is presented as a novel potential biomarker in the progression of disease because of its decreased levels in patients across all BOS grades. Additionally, biomarkers involving the remodeling of the extracellular matrix (ECM), such as MMP-9 and CTSL1, were increased in BOS patients.
Collapse
Affiliation(s)
- Anna Niroomand
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Haider Ghaidan
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 221 85, Lund, Sweden
| | - Oskar Hallgren
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Lennart Hansson
- Department of Pulmonology and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Hillevi Larsson
- Department of Pulmonology and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Darcy Wagner
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Experimental Medical Sciences, Lung Bioengineering and Regeneration, Lund University, Lund, Sweden
| | - Martina Mackova
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Kieran Halloran
- Alberta Transplant Applied Genomics Center, University of Alberta, Edmonton, Canada
| | - Snejana Hyllén
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden. .,Department of Clinical Sciences, Lund University, Lund, Sweden. .,Lund Stem Cell Center, Lund University, Lund, Sweden. .,Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 221 85, Lund, Sweden.
| |
Collapse
|
27
|
Ravichandran R, Bansal S, Rahman M, Sureshbabu A, Sankpal N, Fleming T, Bharat A, Mohanakumar T. Extracellular Vesicles Mediate Immune Responses to Tissue-Associated Self-Antigens: Role in Solid Organ Transplantations. Front Immunol 2022; 13:861583. [PMID: 35572510 PMCID: PMC9094427 DOI: 10.3389/fimmu.2022.861583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Transplantation is a treatment option for patients diagnosed with end-stage organ diseases; however, long-term graft survival is affected by rejection of the transplanted organ by immune and nonimmune responses. Several studies have demonstrated that both acute and chronic rejection can occur after transplantation of kidney, heart, and lungs. A strong correlation has been reported between de novo synthesis of donor-specific antibodies (HLA-DSAs) and development of both acute and chronic rejection; however, some transplant recipients with chronic rejection do not have detectable HLA-DSAs. Studies of sera from such patients demonstrate that immune responses to tissue-associated antigens (TaAgs) may also play an important role in the development of chronic rejection, either alone or in combination with HLA-DSAs. The synergistic effect between HLA-DSAs and antibodies to TaAgs is being established, but the underlying mechanism is yet to be defined. We hypothesize that HLA-DSAs damage the transplanted donor organ resulting in stress and leading to the release of extracellular vesicles, which contribute to chronic rejection. These vesicles express both donor human leukocyte antigen (HLA) and non-HLA TaAgs, which can activate antigen-presenting cells and lead to immune responses and development of antibodies to both donor HLA and non-HLA tissue-associated Ags. Extracellular vesicles (EVs) are released by cells under many circumstances due to both physiological and pathological conditions. Primarily employing clinical specimens obtained from human lung transplant recipients undergoing acute or chronic rejection, our group has demonstrated that circulating extracellular vesicles display both mismatched donor HLA molecules and lung-associated Ags (collagen-V and K-alpha 1 tubulin). This review focuses on recent studies demonstrating an important role of antibodies to tissue-associated Ags in the rejection of transplanted organs, particularly chronic rejection. We will also discuss the important role of extracellular vesicles released from transplanted organs in cross-talk between alloimmunity and autoimmunity to tissue-associated Ags after solid organ transplantation.
Collapse
Affiliation(s)
| | - Sandhya Bansal
- 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
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Narendra Sankpal
- 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
| | - Ankit Bharat
- Department of Surgery-Thoracic, Northwestern University, Chicago, IL, United States
| | | |
Collapse
|
28
|
Magnusson JM, Ricksten A, Dellgren G, Wasslavik C, Nordén R, Westin J, Boehmer J. Cell‐free DNA as a biomarker after lung transplantation: A proof‐of‐concept study. Immun Inflamm Dis 2022; 10:e620. [PMID: 35478446 PMCID: PMC9017613 DOI: 10.1002/iid3.620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 02/28/2022] [Accepted: 03/21/2022] [Indexed: 11/12/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Jesper M. Magnusson
- Transplant Institute Sahlgrenska University Hospital Gothenburg Sweden
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Anne Ricksten
- Department of Clinical Chemistry Sahlgrenska University Hospital Gothenburg Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Göran Dellgren
- Transplant Institute Sahlgrenska University Hospital Gothenburg Sweden
- Department of Cardiothoracic Surgery, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Carina Wasslavik
- Department of Clinical Chemistry Sahlgrenska University Hospital Gothenburg Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Rickard Nordén
- Department of Clinical Microbiology Sahlgrenska University Hospital, Region Västra Götaland Gothenburg Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Johan Westin
- Department of Clinical Microbiology Sahlgrenska University Hospital, Region Västra Götaland Gothenburg Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Jens Boehmer
- Department of Pediatrics, Queen Silvias Children's Hospital Sahlgrenska University Hospital Gothenburg Sweden
- Department of Cardiology, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| |
Collapse
|
29
|
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
|
30
|
Kotecha S, Ivulich S, Snell G. Review: immunosuppression for the lung transplant patient. J Thorac Dis 2022; 13:6628-6644. [PMID: 34992841 PMCID: PMC8662512 DOI: 10.21037/jtd-2021-11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Lung transplantation (LTx) has evolved significantly since its inception and the improvement in LTx outcomes over the last three decades has predominantly been driven by advances in immunosuppression management. Despite the lack of new classes of immunosuppression medications, immunosuppressive strategies have evolved significantly from a universal method to a more targeted approach, reflecting a greater understanding of the need for individualized therapy and careful consideration of all factors that are influenced by immunosuppression choice. This has become increasingly important as the demographics of lung transplant recipients have changed over time, with older and more medically complex candidates being accepted and undergoing LTx. Furthermore, improved survival post lung transplant has translated into more immunosuppression related comorbidities long-term, predominantly chronic kidney disease (CKD) and malignancy, which has required further nuanced management approaches. This review provides an update on current traditional lung transplant immunosuppression strategies, with modifications based on pre-existing recipient factors and comorbidities, peri-operative challenges and long term complications, balanced against the perpetual challenge of chronic lung allograft dysfunction (CLAD). As we continue to explore and understand the complexity of LTx immunology and the interplay of different factors, immunosuppression strategies will require ongoing critical evaluation and personalization in order to continue to improve lung transplant outcomes.
Collapse
Affiliation(s)
- Sakhee Kotecha
- Lung Transplant Service, Alfred Hospital and Monash University, Melbourne, Australia
| | - Steven Ivulich
- Lung Transplant Service, Alfred Hospital and Monash University, Melbourne, Australia
| | - Gregory Snell
- Lung Transplant Service, Alfred Hospital and Monash University, Melbourne, Australia
| |
Collapse
|
31
|
Ravichandran R, Itabashi Y, Liu W, Bansal S, Rahman M, Poulson C, Fleming T, Bremner RM, Smith M, Mohanakumar T. A decline in club cell secretory proteins in lung transplantation is associated with release of natural killer cells exosomes leading to chronic rejection. J Heart Lung Transplant 2021; 40:1517-1528. [PMID: 34627707 PMCID: PMC11019779 DOI: 10.1016/j.healun.2021.08.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/06/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND In human lung transplant recipients, a decline in club cell secretory protein (CCSP) in bronchoalveolar lavage fluid has been associated with chronic lung allograft dysfunction (CLAD) as well as the induction of exosomes and immune responses that lead to CLAD. However, the mechanisms by which CCSP decline contributes to CLAD remain unknown. METHODS To define the mechanisms leading to CCSP decline and chronic rejection, we employed two mouse models: 1) chronic rejection after orthotopic single lung transplantation and 2) anti-major histocompatibility complex (MHC) class I-induced obliterative airway disease. RESULTS In the chronic rejection mouse model, we detected circulating exosomes with donor MHC (H2b) and lung self-antigens and also development of antibodies to H2b and lung self-antigens and then a decline in CCSP. Furthermore, DBA2 mice that received injections of these exosomes developed antibodies to donor MHC and lung self-antigens. In the chronic rejection mouse model, natural killer (NK) and CD8 T cells were the predominant graft-infiltrating cells on day 14 of rejection followed by exosomes containing NK cell-associated and cytotoxic molecules on day 14 and 28. When NK cells were depleted, exosomes with NK cell-associated and cytotoxic molecules as well as fibrosis decreased. CONCLUSIONS Induction of exosomes led to immune responses to donor MHC and lung self-antigens, resulting in CCSP decline, leading to NK cell infiltration and release of exosomes from NK cells. These results suggest a novel role for exosomes derived from NK cells in the pathogenesis of chronic lung allograft rejection.
Collapse
Affiliation(s)
| | | | - Wei Liu
- Norton Thoracic Institute, Phoenix, Arizona
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Sharma AK. The emerging role of exosomal cargo in allograft rejection after lung transplantation. J Heart Lung Transplant 2021; 41:34-36. [PMID: 34756654 DOI: 10.1016/j.healun.2021.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022] Open
Affiliation(s)
- Ashish K Sharma
- Departments of Surgery, Pulmonary & Critical Care Medicine, Physiology & Functional Genomics, and Pharmacology & Therapeutics, University of Florida, Gainesville, Florida.
| |
Collapse
|
33
|
Shevchenko O, Tsirulnikova O, Sharapchenko S, Pashkov I, Bekov M, Shigaev E, Gichkun O, Velikiy D, Gautier S. MiR-339 and galectin-3: diagnostic value in patients with airway obstruction after lung transplantation. Transpl Int 2021; 34:1733-1739. [PMID: 34448266 DOI: 10.1111/tri.13986] [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: 07/13/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Respiratory complications can be the cause of graft dysfunction after lung transplantation (LTx). MicroRNAs are small regulatory molecules-potential biomarkers of respiratory diseases and post-transplant complications. Galectin-3 is highly expressed in fibrosis of transplanted solid organs. The aim was to evaluate the expression of plasma miR-339 and galectin-3 concentrations in lung recipients including with airway obstruction after LTx. The study included 57 lung recipients (34 men and 23 women aged 10 to 74 years) were followed up to 5 years after LTx. The plasma microRNAs were detected by real-time PCR; galectin-3 levels were measured by ELISA. During follow-up in 30 recipients, post-transplant complications were detected: 12 (40.0%) cases of airway obstruction. The levels of miR-339 and galectin-3 were significantly higher in recipients with airway obstruction compare with 27 (47.3%) recipients without any complications (P = 0.036 and P = 0.014, resp.). Increasing miR-339 (above the 0.02 fold change) and galectin-3 (above the 11.7 ng/ml) threshold plasma levels in lung recipients is associated with high risk (RR = 7.14 ± 0.97 [95% CI 1.05-48.60], P = 0.045) of airway obstruction after LTx. A measurement of miR-339 expression in combination with galectin-3 level might be perspective to identify recipients at risk of airway obstruction after LTx.
Collapse
Affiliation(s)
- Olga Shevchenko
- Department of Regulatory mechanisms in Transplantology, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation.,Department of Transplantology and Artificial Organs, Sechenov University, Moscow, Russian Federation
| | - Olga Tsirulnikova
- Department of Transplantology and Artificial Organs, Sechenov University, Moscow, Russian Federation.,Department of Therapy, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Sofya Sharapchenko
- Department of Regulatory mechanisms in Transplantology, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Ivan Pashkov
- Department of Surgery №3, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Maksat Bekov
- Department of Endoscopy, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Egor Shigaev
- Department of Surgery №3, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Olga Gichkun
- Department of Regulatory mechanisms in Transplantology, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation.,Department of Transplantology and Artificial Organs, Sechenov University, Moscow, Russian Federation
| | - Dmitriy Velikiy
- Department of Regulatory mechanisms in Transplantology, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| | - Sergey Gautier
- Department of Transplantology and Artificial Organs, Sechenov University, Moscow, Russian Federation.,Department of Surgery №3, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation.,Director, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russian Federation
| |
Collapse
|
34
|
Liposomes Loaded with Everolimus and Coated with Hyaluronic Acid: A Promising Approach for Lung Fibrosis. Int J Mol Sci 2021; 22:ijms22147743. [PMID: 34299359 PMCID: PMC8303794 DOI: 10.3390/ijms22147743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) and interstitial lung disease associated with collagen tissue diseases (CTD-ILD) are two end-stage lung disorders in which different chronic triggers induce activation of myo-/fibroblasts (LFs). Everolimus, an mTOR inhibitor, can be adopted as a potential strategy for CLAD and CTD-ILD, however it exerts important side effects. This study aims to exploit nanomedicine to reduce everolimus side effects encapsulating it inside liposomes targeted against LFs, expressing a high rate of CD44. PEGylated liposomes were modified with high molecular weight hyaluronic acid and loaded with everolimus (PEG-LIP(ev)-HA400kDa). Liposomes were tested by in vitro experiments using LFs derived from broncholveolar lavage (BAL) of patients affected by CLAD and CTD-ILD, and on alveolar macrophages (AM) and lymphocytes isolated, respectively, from BAL and peripheral blood. PEG-LIP-HA400kDa demonstrated to be specific for LFs, but not for CD44-negative cells, and after loading everolimus, PEG-LIP(ev)-HA400kDa were able to arrest cell cycle arrest and to decrease phospho-mTOR level. PEG-LIP(ev)-HA400kDa showed anti-inflammatory effect on immune cells. This study opens the possibility to use everolimus in lung fibrotic diseases, demonstrating that our lipids-based vehicles can vehicle everolimus inside cells exerting the same drug molecular effect, not only in LFs, but also in immune cells.
Collapse
|
35
|
Malone J, Lee AMD, Hohert G, Nador RG, Lane P. Small airway dilation measured by endoscopic optical coherence tomography correlates with chronic lung allograft dysfunction. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210057R. [PMID: 34263577 PMCID: PMC8278781 DOI: 10.1117/1.jbo.26.7.076005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
SIGNIFICANCE Chronic lung allograft dysfunction (CLAD) is the leading cause of death in transplant patients who survive past the first year post-transplant. Current diagnosis is based on sustained decline in lung function; there is a need for tools that can identify CLAD onset. AIM Endoscopic optical coherence tomography (OCT) can visualize structural changes in the small airways, which are of interest in CLAD progression. We aim to identify OCT features in the small airways of lung allografts that correlate with CLAD status. APPROACH Imaging was conducted with an endoscopic rotary pullback OCT catheter during routine bronchoscopy procedures (n = 54), collecting volumetric scans of three segmental airways per patient. Six features of interest were identified, and four blinded raters scored the dataset on the presence and intensity of each feature. RESULTS Airway dilation (AD) was the only feature found to significantly (p < 0.003) correlate with CLAD diagnosis (R = 0.40 to 0.61). AD could also be fairly consistently scored between raters (κinter-rater = 0.48, κintra-rater = 0.64). There is a stronger relationship between AD and the combined obstructive and restrictive (BOS + RAS) phenotypes than the obstructive-only (BOS) phenotype for two raters (R = 0.92 , 0.94). CONCLUSIONS OCT examination of small AD shows potential as a diagnostic indicator for CLAD and CLAD phenotype and merits further exploration.
Collapse
Affiliation(s)
- Jeanie Malone
- British Columbia Cancer Research Institute, Department of Integrative Oncology, Imaging Unit, Vancouver, BC, Canada
| | - Anthony M. D. Lee
- British Columbia Cancer Research Institute, Department of Integrative Oncology, Imaging Unit, Vancouver, BC, Canada
| | - Geoffrey Hohert
- British Columbia Cancer Research Institute, Department of Integrative Oncology, Imaging Unit, Vancouver, BC, Canada
| | - Roland G. Nador
- University of British Columbia, Division of Respiratory Medicine, Faculty of Medicine, Vancouver, BC, Canada
- Vancouver General Hospital, Lung Transplant Program, Vancouver, BC, Canada
| | - Pierre Lane
- British Columbia Cancer Research Institute, Department of Integrative Oncology, Imaging Unit, Vancouver, BC, Canada
| |
Collapse
|
36
|
Nemska S, Daubeuf F, Obrecht A, Israel-Biet D, Stern M, Kessler R, Roux A, Tavakoli R, Villa P, Tissot A, Danger R, Reber L, Durand E, Foureau A, Brouard S, Magnan A, Frossard N. Overexpression of the MSK1 Kinase in Patients With Chronic Lung Allograft Dysfunction and Its Confirmed Role in a Murine Model. Transplantation 2021; 105:1212-1224. [PMID: 33560725 DOI: 10.1097/tp.0000000000003606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) and its obstructive form, the obliterative bronchiolitis (OB), are the main long-term complications related to high mortality rate postlung transplantation. CLAD treatment lacks a significant success in survival. Here, we investigated a new strategy through inhibition of the proinflammatory mitogen- and stress-activated kinase 1 (MSK1) kinase. METHODS MSK1 expression was assessed in a mouse OB model after heterotopic tracheal allotransplantation. Pharmacological inhibition of MSK1 (H89, fasudil, PHA767491) was evaluated in the murine model and in a translational model using human lung primary fibroblasts in proinflammatory conditions. MSK1 expression was graded over time in biopsies from a cohort of CLAD patients. RESULTS MSK1 mRNA progressively increased during OB (6.4-fold at D21 posttransplantation). Inhibition of MSK1 allowed to counteract the damage to the epithelium (56% restoration for H89), and abolished the recruitment of MHCII+ (94%) and T cells (100%) at the early inflammatory phase of OB. In addition, it markedly decreased the late fibroproliferative obstruction in allografts (48%). MSK1 inhibitors decreased production of IL-6 (whose transcription is under the control of MSK1) released from human lung fibroblasts (96%). Finally, we confirmed occurrence of a 2.9-fold increased MSK1 mRNA expression in lung biopsies in patients at 6 months before CLAD diagnosis as compared to recipients with stable lung function. CONCLUSIONS These findings suggest the overall interest of the MSK1 kinase either as a marker or as a potential therapeutic target in lung dysfunction posttransplantation.
Collapse
Affiliation(s)
- Simona Nemska
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | - Adeline Obrecht
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | | | - Marc Stern
- Hôpital Foch, Suresnes, INRAe UMR 0892, Université de Versailles Saint-Quentin Paris-Saclay, Paris, France
| | - Romain Kessler
- Service de Pneumologie, CHU Strasbourg, Strasbourg, France
| | - Antoine Roux
- Hôpital Foch, Suresnes, INRAe UMR 0892, Université de Versailles Saint-Quentin Paris-Saclay, Paris, France
| | - Reza Tavakoli
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Pascal Villa
- Plateforme de Chimie Biologie Intégrative de Strasbourg (PCBIS) UMS 3286 CNRS, Université de Strasbourg, Labex Medalis, 300 Bld Brant, Illkirch, France
| | - Adrien Tissot
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Richard Danger
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| | - Laurent Reber
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Eugénie Durand
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
| | - Aurore Foureau
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Sophie Brouard
- CHU Nantes, Inserm, UMR 1064, Centre de Recherche en Transplantation et Immunologie, Nantes Université, ITUN, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| | - Antoine Magnan
- Service de Pneumologie, L'institut du thorax, CHU Nantes, Nantes, France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique UMR 7200, LabEx Medalis, CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| |
Collapse
|
37
|
Vanstapel A, Goldschmeding R, Broekhuizen R, Nguyen T, Sacreas A, Kaes J, Heigl T, Verleden SE, De Zutter A, Verleden G, Weynand B, Verbeken E, Ceulemans LJ, Van Raemdonck DE, Neyrinck AP, Schoemans HM, Vanaudenaerde BM, Vos R. Connective Tissue Growth Factor Is Overexpressed in Explant Lung Tissue and Broncho-Alveolar Lavage in Transplant-Related Pulmonary Fibrosis. Front Immunol 2021; 12:661761. [PMID: 34122421 PMCID: PMC8187127 DOI: 10.3389/fimmu.2021.661761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022] Open
Abstract
Background Connective tissue growth factor (CTGF) is an important mediator in several fibrotic diseases, including lung fibrosis. We investigated CTGF-expression in chronic lung allograft dysfunction (CLAD) and pulmonary graft-versus-host disease (GVHD). Materials and Methods CTGF expression was assessed by quantitative real-time polymerase chain reaction (qPCR) and immunohistochemistry in end-stage CLAD explant lung tissue (bronchiolitis obliterans syndrome (BOS), n=20; restrictive allograft syndrome (RAS), n=20), pulmonary GHVD (n=9). Unused donor lungs served as control group (n=20). Next, 60 matched lung transplant recipients (BOS, n=20; RAS, n=20; stable lung transplant recipients, n=20) were included for analysis of CTGF protein levels in plasma and broncho-alveolar lavage (BAL) fluid at 3 months post-transplant, 1 year post-transplant, at CLAD diagnosis or 2 years post-transplant in stable patients. Results qPCR revealed an overall significant difference in the relative content of CTGF mRNA in BOS, RAS and pulmonary GVHD vs. controls (p=0.014). Immunohistochemistry showed a significant higher percentage and intensity of CTGF-positive respiratory epithelial cells in BOS, RAS and pulmonary GVHD patients vs. controls (p<0.0001). BAL CTGF protein levels were significantly higher at 3 months post-transplant in future RAS vs. stable or BOS (p=0.028). At CLAD diagnosis, BAL protein content was significantly increased in RAS patients vs. stable (p=0.0007) and BOS patients (p=0.042). CTGF plasma values were similar in BOS, RAS, and stable patients (p=0.74). Conclusions Lung CTGF-expression is increased in end-stage CLAD and pulmonary GVHD; and higher CTGF-levels are present in BAL of RAS patients at CLAD diagnosis. Our results suggest a potential role for CTGF in CLAD, especially RAS, and pulmonary GVHD.
Collapse
Affiliation(s)
- Arno Vanstapel
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Annelore Sacreas
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium
| | - Janne Kaes
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium
| | - Tobias Heigl
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium
| | - Stijn E Verleden
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium
| | - Alexandra De Zutter
- Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit, Leuven, Belgium
| | - Geert Verleden
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Respiratory Diseases, Lung Transplant Unit, University Hospital Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Erik Verbeken
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Thoracic Surgery University Hospital Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Thoracic Surgery University Hospital Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Cardiovascular Sciences, Katholieke Universiteit, Leuven, Belgium.,Department of Anesthesiology, University Hospital Leuven, Leuven, Belgium
| | | | - Bart M Vanaudenaerde
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, Katholieke Universiteit, Leuven, Belgium.,Department of Respiratory Diseases, Lung Transplant Unit, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Hoang-Thi TN, Chassagnon G, Hua-Huy T, Boussaud V, Dinh-Xuan AT, Revel MP. Chronic Lung Allograft Dysfunction Post Lung Transplantation: A Review of Computed Tomography Quantitative Methods for Detection and Follow-Up. J Clin Med 2021; 10:jcm10081608. [PMID: 33920108 PMCID: PMC8069908 DOI: 10.3390/jcm10081608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/27/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) remains the leading cause of morbidity and mortality after lung transplantation. The term encompasses both obstructive and restrictive phenotypes, as well as mixed and undefined phenotypes. Imaging, in addition to pulmonary function tests, plays a major role in identifying the CLAD phenotype and is essential for follow-up after lung transplantation. Quantitative imaging allows for the performing of reader-independent precise evaluation of CT examinations. In this review article, we will discuss the role of quantitative imaging methods for evaluating the airways and the lung parenchyma on computed tomography (CT) images, for an early identification of CLAD and for prognostic estimation. We will also discuss their limits and the need for novel approaches to predict, understand, and identify CLAD in its early stages.
Collapse
Affiliation(s)
- Trieu-Nghi Hoang-Thi
- AP-HP.Centre, Hôpital Cochin, Department of Radiology, Université de Paris, 75014 Paris, France; (T.-N.H.-T.); (G.C.)
- Department of Diagnostic Imaging, Vinmec Central Park Hospital, Ho Chi Minh City 70000, Vietnam
- AP-HP.Centre, Hôpital Cochin, Department of Respiratory Physiology, Université de Paris, 75014 Paris, France; (T.H.-H.); (A.-T.D.-X.)
| | - Guillaume Chassagnon
- AP-HP.Centre, Hôpital Cochin, Department of Radiology, Université de Paris, 75014 Paris, France; (T.-N.H.-T.); (G.C.)
| | - Thong Hua-Huy
- AP-HP.Centre, Hôpital Cochin, Department of Respiratory Physiology, Université de Paris, 75014 Paris, France; (T.H.-H.); (A.-T.D.-X.)
| | - Veronique Boussaud
- AP-HP.Centre, Hôpital Cochin, Department of Pneumology, Université de Paris, 75014 Paris, France;
| | - Anh-Tuan Dinh-Xuan
- AP-HP.Centre, Hôpital Cochin, Department of Respiratory Physiology, Université de Paris, 75014 Paris, France; (T.H.-H.); (A.-T.D.-X.)
| | - Marie-Pierre Revel
- AP-HP.Centre, Hôpital Cochin, Department of Radiology, Université de Paris, 75014 Paris, France; (T.-N.H.-T.); (G.C.)
- Correspondence: ; Tel.: +33-1-5841-2471
| |
Collapse
|
39
|
Mohanka M, Pinho DF, Garcia H, Kanade R, Bollineni S, Joerns J, Kaza V, Mathews D, Torres F, Zhang S, Banga A. Spectrum of findings on ventilation‒perfusion lung scintigraphy after lung transplantation and association with outcomes. J Heart Lung Transplant 2021; 40:377-386. [PMID: 33648871 DOI: 10.1016/j.healun.2021.01.007] [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: 07/15/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Air trapping (AT) is one of the hallmarks of allograft dysfunction after lung transplantation (LT). Inert gas‒based ventilation‒perfusion (VQ) lung scintigraphy has excellent sensitivity in the detection of AT. METHODS We reviewed the charts of patients who underwent single or double LT between January 2012 and December 2014 (N = 193). Patients without a VQ scintigraphy at the first annual visit (n = 16) and those who did not survive till 1 year (n = 26) were excluded (final n = 151, mean age = 55.8 [SD =14] years, male = 85, female = 66). VQ scintigraphy was independently reviewed and reconciled for the presence and severity of AT by 2 investigators blinded to the clinical data (D.F.P. and D.M.). A 3-year post-transplant survival was the primary end-point. RESULTS AT was common (n = 73, 48.3%). Patients with obstructive lung diseases as the underlying diagnosis (adjusted odds ratio [OR], 4.36, 95% CI: 1.64‒11.6; p = 0.003) and those with lower body mass index (BMI) (BMI < 25 kg/m2 and 25‒30 kg/m2; p < 0.001) had an increased risk of developing AT in the allograft. The presence of AT (adjusted OR, 2.33, 95% CI: 1.01‒5.36; p = 0.04) and peak forced expiratory volume in 1 sec (FEV1) <60% predicted during the first year after LT were independently associated with 3-year mortality. The association of AT with post-transplant mortality was the strongest among patients with BMI <30 kg/m2 and peak FEV1 <60% predicted. CONCLUSIONS The finding of AT on VQ scintigraphy at the first annual visit after LT is independently associated with worse post-transplant mortality. The sub-group of patients who fail to achieve a peak FEV1 of 60% predicted during the first year after LT appears to be the key driver of this association.
Collapse
Affiliation(s)
- Manish Mohanka
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daniella F Pinho
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Heriberto Garcia
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rohan Kanade
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Srinivas Bollineni
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John Joerns
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vaidehi Kaza
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dana Mathews
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Fernando Torres
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Song Zhang
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Amit Banga
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
40
|
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
|
41
|
Sanders YY. New Clue: Prediction from Cell-Free DNA. J Clin Med 2020; 9:jcm9072307. [PMID: 32708076 PMCID: PMC7408910 DOI: 10.3390/jcm9072307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 01/05/2023] Open
Abstract
The main challenge for a positive long-term outcome in lung transplantation is the lack of early detection for chronic lung allograft dysfunction (CLAD). With advancements in technology, an increasing number of studies demonstrate that cell-free DNA (cfDNA) in body fluids could be used as a marker for disease diagnosis, prognosis or monitoring response to treatment. A previous report from this journal found the joint assessment of cfDNA and CXCL10 from brochoalveolar lavage (BAL) could determine the subphenotypes of CLAD and predict lung transplant survival. This is an exciting attempt in monitoring the progress for lung transplant recipients. More studies and better understanding of cfDNA are needed to develop an accessible and reliable biomarker to monitor the progress of CLAD to improve the long-term survival for lung transplant recipients.
Collapse
Affiliation(s)
- Yan Y Sanders
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
42
|
Courtwright AM, Lamattina AM, Takahashi M, Trindade AJ, Hunninghake GM, Rosas IO, Agarwal S, Raby BA, Goldberg HJ, El-Chemaly S. Shorter telomere length following lung transplantation is associated with clinically significant leukopenia and decreased chronic lung allograft dysfunction-free survival. ERJ Open Res 2020; 6:00003-2020. [PMID: 32577419 PMCID: PMC7293991 DOI: 10.1183/23120541.00003-2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/26/2020] [Indexed: 01/11/2023] Open
Abstract
Patients with short telomeres and interstitial lung disease may have decreased chronic lung allograft dysfunction (CLAD)-free survival following lung transplantation. The relationship between post-transplant telomere length and outcomes following lung transplantation has not been characterised among all recipients, regardless of native lung disease. This was a single-centre prospective cohort study. Consenting transplant recipients had their telomere length measured using quantitative real-time PCR assays on peripheral blood collected at the time of surveillance bronchoscopy. We assessed the association between early post-transplant telomere length (as measured in the first 100 days) and CLAD-free survival, time to clinically significant leukopenia, cytomegalovirus (CMV) viraemia, chronic kidney disease, and acute cellular rejection. We also assessed the association between rate of telomere shortening and CLAD-free survival. Telomere lengths were available for 98 out of 215 (45.6%) recipients who underwent lung transplant during the study period (median measurement per patient=2 (interquartile range, 1–3)). Shorter telomere length was associated with decreased CLAD-free survival (hazard ratio (HR)=1.24; 95% CI=1.03–1.48; p=0.02), leukopenia requiring granulocyte colony-stimulating factor (HR=1.17, 95% CI=1.01–1.35, p=0.03), and CMV viraemia among CMV-mismatch recipients (HR=4.04, 95% CI=1.05–15.5, p=0.04). Telomere length was not associated with acute cellular rejection or chronic kidney disease. Recipients with more rapid loss in telomere length (defined as the highest tertile of telomere shortening) did not have worse subsequent CLAD-free survival than those without rapid loss (HR=1.38, 95% CI=0.27–7.01, p=0.70). Shorter early post-transplant telomere length is associated with decreased CLAD-free survival and clinically significant leukopenia in lung transplant recipients, regardless of native lung disease. Shorter recipient telomere length following lung transplantation is associated with clinically significant leukopenia and decreased chronic lung allograft dysfunction-free survivalhttps://bit.ly/2ytymXc
Collapse
Affiliation(s)
- Andrew M Courtwright
- Dept of Pulmonary and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Mai Takahashi
- Harvard T.H. Chen School of Public Health, Boston, MA, USA
| | - Anil J Trindade
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Gary M Hunninghake
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Suneet Agarwal
- Division of Hematology/Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston Children's Hospital, Boston, MA, USA
| | - Benjamin A Raby
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hilary J Goldberg
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
43
|
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
|