1
|
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
|
2
|
Habert P, Chetrit E, Coiffard B, Bregeon F, Thomas P, Loundou A, Bermudez J, Reynaud-Gaubert M, Gaubert JY. Early chest CT abnormalities to predict the subsequent occurrence of chronic lung allograft dysfunction. Insights Imaging 2023; 14:154. [PMID: 37741923 PMCID: PMC10517910 DOI: 10.1186/s13244-023-01509-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/12/2023] [Indexed: 09/25/2023] Open
Abstract
INTRODUCTION Chronic lung allograft dysfunction (CLAD) can take two forms: bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS). The aim was to determine if chest-CT abnormalities after lung transplantation (LTx) could predict CLAD before respiratory functional deterioration. MATERIALS AND METHODS This monocentric retrospective study analyzed consecutive patients who underwent LTx from January 2015 to December 2018. Initial CT post-LTx (CTi) and a follow-up CT at least 9 months post-LTx (CTf) were reviewed. CLAD was defined as a persistent respiratory functional decline (> 20% of basal FEV1) outside acute episode. A Cox regression was performed in univariate, then in multivariate analysis (including features with p < 0.01 in univariate or of clinical importance) to determine risk factors for CLAD. Subgroup analyses were made for BOS, RAS, and death. RESULTS Among 118 LTx patients (median (min-max) 47 (18-68) years), 25 developed CLAD during follow-up (19 BOS). The median time to CLAD since LTx was 570 days [150-1770]. Moderate pulmonary artery stenosis (30-50%) was associated with the occurrence of CLAD on CTi (hazard ratio HR = 4.6, CI [1.6-13.2]) and consolidations and pleural effusion on CTf (HR = 2.6, CI [1.3-4.9] and HR = 4.5, CI [1.5-13.6] respectively). The presence of mosaic attenuation (HR = 4.1, CI [1.4-12.5]), consolidations (HR = 2.6, CI [1.3-5.4]), and pleural effusions (p = 0.01, HR = 5.7, CI [1.4-22.3]) were risk factors for BOS on CTf. The consolidations (p = 0.029) and pleural effusions (p = 0.001) were risk factors for death on CTf. CONCLUSIONS CTi and CTf in the monitoring of LTx patients could predict CLAD. Moderate pulmonary artery stenosis, mosaic pattern, parenchyma condensations, and pleural effusions were risk factors for CLAD. CRITICAL RELEVANCE STATEMENT There is a potential predictive role of chest CT in the follow-up of LTx patients for chronic lung allograft dysfunction (CLAD). Early chest CT should focus on pulmonary artery stenosis (risk factor for CLAD in this study). During the follow-up (at least 9 months post-LTx), parenchymal consolidations and pleural effusions were shown to be risk factors for CLAD, and death in subgroup analyses. KEY POINTS • Pulmonary artery stenosis (30-50%) on initial chest-CT following lung transplantation predicts CLAD HR = 4.5; CI [1.6-13.2]. • Pleural effusion and consolidations 1 year after lung transplantation predict CLAD and death. • Early evaluation of lung transplanted patients should evaluate pulmonary artery anastomosis.
Collapse
Affiliation(s)
- Paul Habert
- Service de radiologie, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France.
- Aix Marseille Univ, LIIE, Marseille, France.
- Aix Marseille Univ, CERIMED, Marseille, France.
| | - Elsa Chetrit
- Service de radiologie, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France
| | - Benjamin Coiffard
- Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, AP-HM Hôpital Nord, 13015, Marseille, France
| | - Fabienne Bregeon
- APHM, Hôpital Nord, Explorations Fonctionnelles Respiratoires, Marseille, France
- Aix Marseille Univ, APHM, Microbes Evolution Phylogeny and Infections (MEPHI), IHU-Méditerranée Infection, Marseille, France
| | - Pascal Thomas
- Service de chirurgie thoracique, Hôpital Nord, chemin des Bourrely, 13015, Marseille, France
| | - Anderson Loundou
- Aix-Marseille Univ, - CEReSS UR3279-Health Service Research and Quality of Life Center, Marseille, France
- Department of Public Health, Assistance Publique - Hôpitaux de Marseille, Marseille, France
| | - Julien Bermudez
- Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, AP-HM Hôpital Nord, 13015, Marseille, France
| | - Martine Reynaud-Gaubert
- Centre de Ressources et de Compétences de la Mucoviscidose (CRCM) Adulte, AP-HM Hôpital Nord, 13015, Marseille, France
| | - Jean-Yves Gaubert
- Aix Marseille Univ, LIIE, Marseille, France
- Aix Marseille Univ, CERIMED, Marseille, France
- Service de radiologie, La Timone Hôpital, 264 rue Saint Pierre, 13005, Marseille, France
| |
Collapse
|
3
|
Banday MM, Rao SB, Shankar S, Khanday MA, Finan J, O'Neill E, Coppolino A, Seyfang A, Kumar A, Rinewalt DE, Goldberg HJ, Woolley A, Mallidi HR, Visner G, Gaggar A, Patel KN, Sharma NS. IL-33 mediates Pseudomonas induced airway fibrogenesis and is associated with CLAD. J Heart Lung Transplant 2023; 42:53-63. [PMID: 37014805 PMCID: PMC10260236 DOI: 10.1016/j.healun.2022.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Long term outcomes of lung transplantation are impacted by the occurrence of chronic lung allograft dysfunction (CLAD). Recent evidence suggests a role for the lung microbiome in the occurrence of CLAD, but the exact mechanisms are not well defined. We hypothesize that the lung microbiome inhibits epithelial autophagic clearance of pro-fibrotic proteins in an IL-33 dependent manner, thereby augmenting fibrogenesis and risk for CLAD. METHODS Autopsy derived CLAD and non-CLAD lungs were collected. IL-33, P62 and LC3 immunofluorescence was performed and assessed using confocal microscopy. Pseudomonas aeruginosa (PsA), Streptococcus Pneumoniae (SP), Prevotella Melaninogenica (PM), recombinant IL-33 or PsA-lipopolysaccharide was co-cultured with primary human bronchial epithelial cells (PBEC) and lung fibroblasts in the presence or absence of IL-33 blockade. Western blot analysis and quantitative reverse transcription (qRT) PCR was performed to evaluate IL-33 expression, autophagy, cytokines and fibroblast differentiation markers. These experiments were repeated after siRNA silencing and upregulation (plasmid vector) of Beclin-1. RESULTS Human CLAD lungs demonstrated markedly increased expression of IL-33 and reduced basal autophagy compared to non-CLAD lungs. Exposure of co-cultured PBECs to PsA, SP induced IL-33, and inhibited PBEC autophagy, while PM elicited no significant response. Further, PsA exposure increased myofibroblast differentiation and collagen formation. IL-33 blockade in these co-cultures recovered Beclin-1, cellular autophagy and attenuated myofibroblast activation in a Beclin-1 dependent manner. CONCLUSION CLAD is associated with increased airway IL-33 expression and reduced basal autophagy. PsA induces a fibrogenic response by inhibiting airway epithelial autophagy in an IL-33 dependent manner.
Collapse
Affiliation(s)
- Mudassir M Banday
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Shruthi Shankar
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | | | - Jon Finan
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Edward O'Neill
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Antonio Coppolino
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andreas Seyfang
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Archit Kumar
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel E Rinewalt
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hilary J Goldberg
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ann Woolley
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hari Reddy Mallidi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gary Visner
- Boston Children's Hospital. Harvard Medical School
| | | | - Kapil N Patel
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Nirmal S Sharma
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Boston VA Medical Center.
| |
Collapse
|
4
|
Fifteen-Year Surveillance of LTR Receiving Pre-Emptive Therapy for CMV Infection: Prevention of CMV Disease and Incidence of CLAD. Microorganisms 2022; 10:microorganisms10122339. [PMID: 36557592 PMCID: PMC9788487 DOI: 10.3390/microorganisms10122339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The efficacy of pre-emptive therapy in the prevention of cytomegalovirus (CMV) disease and the potential association of CMV infection with the occurrence of chronic lung allograft dysfunction (CLAD) was evaluated in 129 lung transplant recipients receiving pre-emptive therapy based on pp65-antigenemia or CMV-DNA in the blood and in the bronchoalveolar lavage. Seventy-one (55%) patients received pre-emptive ganciclovir/valganciclovir (GCV/VGCV) for CMV infection for a median of 28 (9-191) days. Possible CMV disease occurred in six (5%) patients and was healed after the GCV/VGCV therapy. The cumulative incidence of CLAD was 38% and 54% at 5 and 10 years. Acute rejection and CMV load in the blood (but not in the lung) were independent predictors of the occurrence of CLAD. Pre-emptive therapy is highly effective in preventing CMV disease in lung recipients and does not induce a superior incidence of CLAD compared to what reported for other cohorts of patients who received an extended antiviral prophylaxis.
Collapse
|
5
|
Effective Extracorporeal Photopheresis of Patients with Transplantation Induced Acute Intestinal GvHD and Bronchiolitis Obliterans Syndrome. Biomedicines 2022; 10:biomedicines10081887. [PMID: 36009436 PMCID: PMC9405770 DOI: 10.3390/biomedicines10081887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Patients with steroid-refractory intestinal acute graft-versus-host disease (aGvHD) and bronchiolitis obliterans syndrome (BOS) represent a population with a high need for alternative and effective treatment options. Methods: We report real-life data from 18 patients treated with extracorporeal photopheresis (ECP). This cohort consisted of nine patients with steroid-refractory intestinal aGvHD and nine patients with BOS. Results: We document partial or complete clinical response and reduction of symptoms in half of the patients with intestinal acute GvHD and patients with BOS treated ECP. Responding patients tended to stay on treatment longer. In patients with BOS, stabilization of lung function and forced expiratory volume was observed, whereas, less abdominal pain, less diarrhea, and a reduction of systemic corticosteroids were seen in patients with intestinal acute GvHD. Conclusions: ECP might not only abrogate symptoms but also reduce mortality caused by complications from high-dose steroid treatment. Taken together, ECP offers a serious treatment avenue for patients with steroid-refractory intestinal acute GvHD and BOS.
Collapse
|
6
|
Santos J, Calabrese DR, Greenland JR. Lymphocytic Airway Inflammation in Lung Allografts. Front Immunol 2022; 13:908693. [PMID: 35911676 PMCID: PMC9335886 DOI: 10.3389/fimmu.2022.908693] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Lung transplant remains a key therapeutic option for patients with end stage lung disease but short- and long-term survival lag other solid organ transplants. Early ischemia-reperfusion injury in the form of primary graft dysfunction (PGD) and acute cellular rejection are risk factors for chronic lung allograft dysfunction (CLAD), a syndrome of airway and parenchymal fibrosis that is the major barrier to long term survival. An increasing body of research suggests lymphocytic airway inflammation plays a significant role in these important clinical syndromes. Cytotoxic T cells are observed in airway rejection, and transcriptional analysis of airways reveal common cytotoxic gene patterns across solid organ transplant rejection. Natural killer (NK) cells have also been implicated in the early allograft damage response to PGD, acute rejection, cytomegalovirus, and CLAD. This review will examine the roles of lymphocytic airway inflammation across the lifespan of the allograft, including: 1) The contribution of innate lymphocytes to PGD and the impact of PGD on the adaptive immune response. 2) Acute cellular rejection pathologies and the limitations in identifying airway inflammation by transbronchial biopsy. 3) Potentiators of airway inflammation and heterologous immunity, such as respiratory infections, aspiration, and the airway microbiome. 4) Airway contributions to CLAD pathogenesis, including epithelial to mesenchymal transition (EMT), club cell loss, and the evolution from constrictive bronchiolitis to parenchymal fibrosis. 5) Protective mechanisms of fibrosis involving regulatory T cells. In summary, this review will examine our current understanding of the complex interplay between the transplanted airway epithelium, lymphocytic airway infiltration, and rejection pathologies.
Collapse
Affiliation(s)
- Jesse Santos
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
| | - Daniel R. Calabrese
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| | - John R. Greenland
- Department of Medicine University of California, San Francisco, San Francisco, CA, United States
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
7
|
Magnusson JM, Ericson P, Tengvall S, Stockfelt M, Brundin B, Lindén A, Riise GC. Involvement of IL-26 in bronchiolitis obliterans syndrome but not in acute rejection after lung transplantation. Respir Res 2022; 23:108. [PMID: 35501858 PMCID: PMC9063324 DOI: 10.1186/s12931-022-02036-3] [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: 09/27/2021] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The main long-term complication after lung transplantation is bronchiolitis obliterans syndrome (BOS), a deadly condition in which neutrophils may play a critical pathophysiological role. Recent studies show that the cytokine interleukin IL-26 can facilitate neutrophil recruitment in response to pro-inflammatory stimuli in the airways. In this pilot study, we characterized the local involvement of IL-26 during BOS and acute rejection (AR) in human patients. METHOD From a biobank containing bronchoalveolar lavage (BAL) samples from 148 lung transplant recipients (LTR), clinically-matched patient pairs were identified to minimize the influence of clinical confounders. We identified ten pairs (BOS/non-BOS) with BAL samples harvested on three occasions for our longitudinal investigation and 12 pairs of patients with and without AR. The pairs were matched for age, gender, preoperative diagnosis, type of and time after surgery. Extracellular IL-26 protein was quantified in cell-free BAL samples using an enzyme-linked immunosorbent assay. Intracellular IL-26 protein in BAL cells was determined using immunocytochemistry (ICC) and flow cytometry. RESULTS The median extracellular concentration of IL-26 protein was markedly increased in BAL samples from patients with BOS (p < 0.0001) but not in samples from patients with AR. Intracellular IL-26 protein was confirmed in alveolar macrophages and lymphocytes (through ICC and flow cytometry) among BAL cells obtained from BOS patients. CONCLUSIONS Local IL-26 seems to be involved in BOS but not AR, and macrophages as well as lymphocytes constitute cellular sources in this clinical setting. The enhancement of extracellular IL-26 protein in LTRs with BOS warrants further investigation of its potential as a target for diagnosing, monitoring, and treating BOS.
Collapse
Affiliation(s)
- Jesper M Magnusson
- Department of Respiratory Medicine, Institute of Medicine Sahlgrenska Academy at the University of Gothenburg, Bruna stråket 11, 41345, Gothenburg, Sweden.
| | - Petrea Ericson
- Department of Respiratory Medicine, Institute of Medicine Sahlgrenska Academy at the University of Gothenburg, Bruna stråket 11, 41345, Gothenburg, Sweden
| | - Sara Tengvall
- Department of Respiratory Medicine, Institute of Medicine Sahlgrenska Academy at the University of Gothenburg, Bruna stråket 11, 41345, Gothenburg, Sweden.,Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marit Stockfelt
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Bettina Brundin
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Lindén
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Gerdt C Riise
- Department of Respiratory Medicine, Institute of Medicine Sahlgrenska Academy at the University of Gothenburg, Bruna stråket 11, 41345, Gothenburg, Sweden
| |
Collapse
|
8
|
Müller C, Rosmark O, Åhrman E, Brunnström H, Wassilew K, Nybom A, Michaliková B, Larsson H, Eriksson LT, Schultz HH, Perch M, Malmström J, Wigén J, Iversen M, Westergren-Thorsson G. Protein Signatures of Remodeled Airways in Transplanted Lungs with Bronchiolitis Obliterans Syndrome Obtained Using Laser-Capture Microdissection. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1398-1411. [PMID: 34111430 DOI: 10.1016/j.ajpath.2021.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 10/25/2022]
Abstract
Bronchiolitis obliterans syndrome, a common form of chronic lung allograft dysfunction, is the major limitation to long-term survival after lung transplantation. The histologic correlate is progressive, fibrotic occlusion of small airways, obliterative bronchiolitis lesions, which ultimately lead to organ failure. The molecular composition of these lesions is unknown. In this sutdy, the protein composition of the lesions in explanted lungs from four end-stage bronchiolitis obliterans syndrome patients was analyzed using laser-capture microdissection and optimized sample preparation protocols for mass spectrometry. Immunohistochemistry and immunofluorescence were used to determine the spatial distribution of commonly identified proteins on the tissue level, and protein signatures for 14 obliterative bronchiolitis lesions were established. A set of 39 proteins, identified in >75% of lesions, included distinct structural proteins (collagen types IV and VI) and cellular components (actins, vimentin, and tryptase). Each respective lesion exhibited a unique composition of proteins (on average, n = 66 proteins), thereby mirroring the morphologic variation of the lesions. Antibody-based staining confirmed these mass spectrometry-based findings. The 14 analyzed obliterative bronchiolitis lesions showed variations in their protein content, but also common features. This study provides molecular and morphologic insights into the development of chronic rejection after lung transplantation. The protein patterns in the lesions were correlated to pathways of extracellular matrix organization, tissue development, and wound healing processes.
Collapse
Affiliation(s)
- Catharina Müller
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oskar Rosmark
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Emma Åhrman
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden; Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Division of Laboratory Medicine, Department of Genetics and Pathology, Region Skåne, Lund, Sweden
| | - Katharina Wassilew
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Annika Nybom
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Barbora Michaliková
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hillevi Larsson
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Leif T Eriksson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden; Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Hans H Schultz
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jenny Wigén
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Martin Iversen
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
9
|
Sweet SC. Community-Acquired Respiratory Viruses Post-Lung Transplant. Semin Respir Crit Care Med 2021; 42:449-459. [PMID: 34030206 DOI: 10.1055/s-0041-1729172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Survival in lung transplant recipients (LTRs) lags behind heart, liver, and kidney transplant, in part due to the direct and indirect effects of infection. LTRs have increased susceptibility to infection due to the combination of a graft continually exposed to the outside world, multiple mechanisms for impaired mucus clearance, and immunosuppression. Community-acquired respiratory viral infections (CARVs) are common in LTRs. Picornaviruses have roughly 40% cumulative incidence followed by respiratory syncytial virus and coronaviruses. Although single-center retrospective and prospective series implicate CARV in rejection and mortality, conclusive evidence for and well-defined mechanistic links to long-term outcome are lacking. Treatment of viral infections can be challenging except for influenza. Future studies are needed to develop better treatments and clarify the links between CARV and long-term outcomes.
Collapse
Affiliation(s)
- Stuart C Sweet
- Division of Allergy and Pulmonary Medicine, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
10
|
Hanka I, Stamminger T, Ramsperger-Gleixner M, Kuckhahn AV, Müller R, Weyand M, Heim C. Role of CMV chemokine receptor M33 in airway graft rejection in a mouse transplant model. Transpl Immunol 2021; 67:101415. [PMID: 34033867 DOI: 10.1016/j.trim.2021.101415] [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: 03/09/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is a risk factor for bronchiolitis obliterans (BO), one form of chronic lung allograft dysfunction (CLAD). The viral chemokine receptor M33 is essential for successful spread of murine CMV to host salivary glands. In the present study we investigated the impact of M33 on chronic airway rejection. METHODS MHC I-mismatched tracheas of C·B10-H2b/LilMcdJ mice were transplanted into BALB/c (H2d) recipients and infected at different dates with wild type (WT) or M33-deleted (delM33) MCMV representing clinical settings of viral recipient (R)-donor (D)-serostatus: (D-/R+) or (D+/R-). Grafts were recovered for gene expression and histological / immunofluorescence analysis, respectively. RESULTS Evaluations showed significantly increased signs of chronic rejection in WT-infected mice compared to uninfected allografts seen in lower epithelium/lamina propria-ratio (ELR) (ELR 0.46 ± 0.07 [WT post] vs. ELR 0.66 ± 0.10 [non-inf.]; p < 0.05). The rejection in delM33-infected groups was significantly reduced vs. WT-infected groups (0.67 ± 0.04 [delM33 post]; vs. WT post p < 0.05). Furthermore, decreased rejection was observed in WT pre-infected compared to post-infected groups (0.56 ± 0.08 [WT pre]; vs. WT post p < 0.05). CD8+ T cell infiltration was significantly higher in WT-post compared to the delM33 infected or non-infected allografts. CONCLUSIONS These data support the role of the CMV in accelerating CLAD. The deletion of chemokine receptor M33 leads to attenuated rejection.
Collapse
Affiliation(s)
- Isabella Hanka
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Thomas Stamminger
- Institute for Virology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Martina Ramsperger-Gleixner
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Annika V Kuckhahn
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Regina Müller
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany.
| |
Collapse
|
11
|
Morrone C, Smirnova NF, Jeridi A, Kneidinger N, Hollauer C, Schupp JC, Kaminski N, Jenne D, Eickelberg O, Yildirim AÖ. Cathepsin B promotes collagen biosynthesis, which drives bronchiolitis obliterans syndrome. Eur Respir J 2021; 57:13993003.01416-2020. [PMID: 33303550 DOI: 10.1183/13993003.01416-2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 11/08/2020] [Indexed: 12/27/2022]
Abstract
Bronchiolitis obliterans syndrome (BOS) is a major complication after lung transplantation (LTx). BOS is characterised by massive peribronchial fibrosis, leading to air trapping-induced pulmonary dysfunction. Cathepsin B, a lysosomal cysteine protease, has been shown to enforce fibrotic pathways in several diseases. However, the relevance of cathepsin B in BOS progression has not yet been addressed. The aim of the study was to elucidate the function of cathepsin B in BOS pathogenesis.We determined cathepsin B levels in bronchoalveolar lavage fluid (BALF) and lung tissue from healthy donors (HD) and BOS LTx patients. Cathepsin B activity was assessed via a fluorescence resonance energy transfer-based assay and protein expression was determined using Western blotting, ELISA and immunostaining. To investigate the impact of cathepsin B in the pathophysiology of BOS, we used an in vivo orthotopic left LTx mouse model. Mechanistic studies were performed in vitro using macrophage and fibroblast cell lines.We found a significant increase of cathepsin B activity in BALF and lung tissue from BOS patients, as well as in our murine model of lymphocytic bronchiolitis. Moreover, cathepsin B activity was associated with increased biosynthesis of collagen and had a negative effect on lung function. We observed that cathepsin B was mainly expressed in macrophages that infiltrated areas characterised by a massive accumulation of collagen deposition. Mechanistically, macrophage-derived cathepsin B contributed to transforming growth factor-β1-dependent activation of fibroblasts, and its inhibition reversed the phenotype.Infiltrating macrophages release active cathepsin B, thereby promoting fibroblast activation and subsequent collagen deposition, which drive BOS. Cathepsin B represents a promising therapeutic target to prevent the progression of BOS.
Collapse
Affiliation(s)
- Carmela Morrone
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Natalia F Smirnova
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Aicha Jeridi
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nikolaus Kneidinger
- Dept of Internal Medicine V, Ludwig Maximilians University of Munich, Munich, Germany.,Comprehensive Pneumology Center, Ludwig Maximilians University of Munich; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christine Hollauer
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jonas Christian Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Dieter Jenne
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.,Max Planck Institute of Neurobiology, Munich, Germany
| | - Oliver Eickelberg
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Pittsburgh, Pittsburg, PA, USA.,Contributed equally to this article as lead authors and supervised the work
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany .,Contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
12
|
Banday MM, Kumar A, Vestal G, Sethi J, Patel KN, O'Neill EB, Finan J, Cheng F, Lin M, Davis NM, Goldberg H, Coppolino A, Mallidi HR, Dunning J, Visner G, Gaggar A, Seyfang A, Sharma NS. N-myc-interactor mediates microbiome induced epithelial to mesenchymal transition and is associated with chronic lung allograft dysfunction. J Heart Lung Transplant 2021; 40:447-457. [PMID: 33781665 DOI: 10.1016/j.healun.2021.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/09/2021] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Recent evidence suggests a role for lung microbiome in occurrence of chronic lung allograft dysfunction (CLAD). However, the mechanisms linking the microbiome to CLAD are poorly delineated. We investigated a possible mechanism involved in microbial modulation of mucosal response leading to CLAD with the hypothesis that a Proteobacteria dominant lung microbiome would inhibit N-myc-interactor (NMI) expression and induce epithelial to mesenchymal transition (EMT). METHODS Explant CLAD, non-CLAD, and healthy nontransplant lung tissue were collected, as well as bronchoalveolar lavage from 14 CLAD and matched non-CLAD subjects, which were followed by 16S rRNA amplicon sequencing and quantitative polymerase chain reaction (PCR) analysis. Pseudomonas aeruginosa (PsA) or PsA-lipopolysaccharide was cocultured with primary human bronchial epithelial cells (PBEC). Western blot analysis and quantitative reverse transcription (qRT) PCR was performed to evaluate NMI expression and EMT in explants and in PsA-exposed PBECs. These experiments were repeated after siRNA silencing and upregulation (plasmid vector) of EMT regulator NMI. RESULTS 16S rRNA amplicon analyses revealed that CLAD patients have a higher abundance of phyla Proteobacteria and reduced abundance of the phyla Bacteroidetes. At the genera level, CLAD subjects had an increased abundance of genera Pseudomonas and reduced Prevotella. Human CLAD airway cells showed a downregulation of the N-myc-interactor gene and presence of EMT. Furthermore, exposure of human primary bronchial epithelial cells to PsA resulted in downregulation of NMI and induction of an EMT phenotype while NMI upregulation resulted in attenuation of this PsA-induced EMT response. CONCLUSIONS CLAD is associated with increased bacterial biomass and a Proteobacteria enriched airway microbiome and EMT. Proteobacteria such as PsA induces EMT in human bronchial epithelial cells via NMI, demonstrating a newly uncovered mechanism by which the microbiome induces cellular metaplasia.
Collapse
Affiliation(s)
- Mudassir M Banday
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Archit Kumar
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Grant Vestal
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Jaskaran Sethi
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Kapil N Patel
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Edward B O'Neill
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Jon Finan
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Feng Cheng
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Muling Lin
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Nicole M Davis
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Hilary Goldberg
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Antonio Coppolino
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hari R Mallidi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - John Dunning
- University of South Florida/Tampa General Hospital,Tampa, Florida
| | - Gary Visner
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amit Gaggar
- University of Alabama at Birmingham, Birmingham, Alabama
| | - Andreas Seyfang
- University of South Florida Morsani College of Medicine/Molecular Medicine, Tampa, Florida
| | - Nirmal S Sharma
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
13
|
Sharma M, Gunasekaran M, Ravichandran R, Fisher CE, Limaye AP, Hu C, McDyer J, Kaza V, Bharat A, Tokman S, Omar A, Arjuna A, Walia R, Bremner RM, Smith MA, Hachem RR, Mohanakumar T. Circulating exosomes with lung self-antigens as a biomarker for chronic lung allograft dysfunction: A retrospective analysis. J Heart Lung Transplant 2020; 39:1210-1219. [PMID: 32713614 DOI: 10.1016/j.healun.2020.07.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Exosomes isolated from plasma of lung transplant recipients (LTxRs) with bronchiolitis obliterans syndrome (BOS) contain human leukocyte antigens and lung self-antigens (SAgs), K-alpha 1 tubulin (Kα1T) and collagen type V (Col-V). The aim was to determine the use of circulating exosomes with lung SAgs as a biomarker for BOS. METHODS Circulating exosomes were isolated retrospectively from plasma from LTxRs at diagnosis of BOS and at 6 and 12 months before the diagnosis (n = 41) and from stable time-matched controls (n = 30) at 2 transplant centers by ultracentrifugation. Exosomes were validated using Nanosight, and lung SAgs (Kα1T and Col-V) were detected by immunoblot and semiquantitated using ImageJ software. RESULTS Circulating exosomes from BOS and stable LTxRs demonstrated 61- to 181-nm vesicles with markers Alix and CD9. Exosomes from LTxRs with BOS (n = 21) showed increased levels of lung SAgs compared with stable (n = 10). A validation study using 2 separate cohorts of LTxRs with BOS and stable time-matched controls from 2 centers also demonstrated significantly increased lung SAgs-containing exosomes at 6 and 12 months before BOS. CONCLUSIONS Circulating exosomes isolated from LTxRs with BOS demonstrated increased levels of lung SAgs (Kα1T and Col-V) 12 months before the diagnosis (100% specificity and 90% sensitivity), indicating that circulating exosomes with lung SAgs can be used as a non-invasive biomarker for identifying LTxRs at risk for BOS.
Collapse
Affiliation(s)
- Monal Sharma
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | | | | | - Cynthia E Fisher
- Deparment of Medicine, University of Washington, Seattle, Washington
| | - Ajit P Limaye
- Deparment of Medicine, University of Washington, Seattle, Washington
| | - Chengcheng Hu
- Department of Epidemiology and Biostatistics, University of Arizona, Phoenix, Arizona
| | - John McDyer
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vaidehi Kaza
- Internal Medicine-Pulmonary Disease, University of Texas Southwestern, Dallas, Texas
| | - Ankit Bharat
- Department of Surgery-Thoracic, Northwestern University, Chicago, Illinois
| | - Sofya Tokman
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Ashraf Omar
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Ashwini Arjuna
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Rajat Walia
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Ross M Bremner
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Michael A Smith
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Ramsey R Hachem
- Department of Internal Medicine, Washington University Medical School, St Louis, Missouri
| | | |
Collapse
|
14
|
Oshima Y, Sato S, Chen-Yoshikawa TF, Yoshioka Y, Shimamura N, Hamada R, Nankaku M, Tamaki A, Date H, Matsuda S. Quantity and quality of antigravity muscles in patients undergoing living-donor lobar lung transplantation: 1-year longitudinal analysis using chest computed tomography images. ERJ Open Res 2020; 6:00205-2019. [PMID: 32665944 PMCID: PMC7335834 DOI: 10.1183/23120541.00205-2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/22/2020] [Indexed: 01/07/2023] Open
Abstract
Background Skeletal muscle dysfunction is a common feature in patients with severe lung diseases. Although lung transplantation aims to save these patients, the surgical procedure and disuse may cause additional deterioration and prolonged functional disability. We investigated the postoperative course of antigravity muscle condition in terms of quantity and quality using chest computed tomography. Methods 35 consecutive patients were investigated for 12 months after living-donor lobar lung transplantation (LDLLT). The erector spinae muscles (ESMs), which are antigravity muscles, were evaluated, and the cross-sectional area (ESMCSA) and mean attenuation (ESMCT) were analysed to determine the quantity and quality of ESMs. Functional capacity was evaluated by the 6-min walk distance (6MWD). Age-matched living donors with lower lobectomy were evaluated as controls. Results Recipient and donor ESMCSA values temporarily decreased at 3 months and recovered by 12 months post-operatively. The ESMCSA of recipients, but not that of donors, surpassed baseline values by 12 months post-operatively. Increased ESMCSA (ratio to baseline ≥1) may occur at 12 months in patients with a high baseline ESMCT. Although the recipient ESMCT may continuously decrease for 12 months, the ESMCT is a major determinant, in addition to lung function, of the postoperative 6MWD at both 3 and 12 months. Conclusion The quantity of ESMs may increase within 12 months after LDLLT in recipients with better muscle quality at baseline. The quality of ESMs is also important for physical performance; therefore, further approaches to prevent deterioration in muscle quality are required. The quantity of antigravity muscles in patients undergoing lung transplantation (LTx) will increase within 1 year after LTx. The quality of muscles is important for increase of muscle quantity as well as physical performance.https://bit.ly/3bItfB9
Collapse
Affiliation(s)
- Yohei Oshima
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| | - Susumu Sato
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan.,Dept of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Yuji Yoshioka
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| | - Nana Shimamura
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| | - Ryota Hamada
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| | - Manabu Nankaku
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| | - Akira Tamaki
- Dept of Rehabilitation Science, Graduate School of Health Science, Hyogo University of Health Sciences, Kobe, Japan
| | - Hiroshi Date
- Dept of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
15
|
Metwally AA, Ascoli C, Turturice B, Rani A, Ranjan R, Chen Y, Schott C, Faro A, Ferkol TW, Finn PW, Perkins DL. Pediatric lung transplantation: Dynamics of the microbiome and bronchiolitis obliterans in cystic fibrosis. J Heart Lung Transplant 2020; 39:824-834. [PMID: 32580896 DOI: 10.1016/j.healun.2020.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/02/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Compositional changes in the microbiome are associated with the development of bronchiolitis obliterans (BO) after lung transplantation (LTx) in adults with cystic fibrosis (CF). The association between the lower airway bacterial community and BO after LTx in children with CF remains largely unexplored and is possibly influenced by frequent antibiotic therapy. The objectives of this study were to examine the relationship between bacterial community dynamics and the development of BO and analyze antibiotic resistance trends in children after LTx for CF. METHODS For 3 years from the time of transplant, 12 LTx recipients were followed longitudinally, with 5 subjects developing BO during the study period. A total of 82 longitudinal bronchoalveolar lavage samples were collected during standard of care bronchoscopies. Metagenomic shotgun sequencing was performed on the extracted microbial DNA from bronchoalveolar lavage specimens. Taxonomic profiling was constructed using WEVOTE pipeline. The longitudinal association between development of BO and temporal changes in bacterial diversity and abundance were evaluated with MetaLonDA. The analysis of antibiotic resistance genes was performed with the ARGs-OAP v2.0 pipeline. RESULTS All recipients demonstrated a Proteobacteria-predominant lower airways community. Temporal reduction in bacterial diversity was significantly associated with the development of BO and associated with neutrophilia and antibiotic therapy. Conversely, an increasing abundance of the phylum Actinobacteria and the orders Neisseriales and Pseudonocardiales in the lower airways was significantly associated with resilience to BO. A more diverse bacterial community was related to a higher expression of multidrug resistance genes and increased proteobacterial abundance. CONCLUSIONS Decreased diversity within bacterial communities may suggest a contribution to pediatric lung allograft rejection in CF.
Collapse
Affiliation(s)
- Ahmed A Metwally
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Genetics, Stanford University, Stanford, California
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Benjamin Turturice
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology
| | - Asha Rani
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ravi Ranjan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yang Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Cody Schott
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology
| | - Albert Faro
- Cystic Fibrosis Foundation, Bethesda, Maryland; Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Thomas W Ferkol
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Patricia W Finn
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Departments of Microbiology and Immunology.
| | - David L Perkins
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois; Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Surgery, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
16
|
Combs MP, Xia M, Wheeler DS, Belloli EA, Walker NM, Braeuer RR, Lyu DM, Murray S, Lama VN. Fibroproliferation in chronic lung allograft dysfunction: Association of mesenchymal cells in bronchoalveolar lavage with phenotypes and survival. J Heart Lung Transplant 2020; 39:815-823. [PMID: 32360292 DOI: 10.1016/j.healun.2020.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD), the primary cause of poor outcome after lung transplantation, arises from fibrotic remodeling of the allograft and presents as diverse clinical phenotypes with variable courses. Here, we investigate whether bronchoalveolar lavage (BAL) mesenchymal cell activity at CLAD onset can inform regarding disease phenotype, progression, and survival. METHODS Mesenchymal cell colony-forming units (CFUs) were measured in BAL obtained at CLAD onset (n = 77) and CLAD-free time post-transplant matched controls (n = 77). CFU counts were compared using Wilcoxon's rank-sum test. Cox proportional hazards and restricted means models were utilized to investigate post-CLAD survival. RESULTS Higher mesenchymal CFU counts were noted in BAL at the time of CLAD onset than in CLAD-free controls. Patients with restrictive allograft syndrome had higher BAL mesenchymal CFU count at CLAD onset than patients with bronchiolitis obliterans syndrome (p = 0.011). Patients with high mesenchymal CFU counts (≥10) at CLAD onset had worse outcomes than those with low (<10) CFU counts, with shorter average survival (2.64 years vs 4.25 years; p = 0.027) and shorter progression-free survival, defined as time to developing either CLAD Stage 3 or death (0.97 years vs 2.70 years; p < 0.001). High CFU count remained predictive of decreased overall survival and progression-free survival after accounting for the CLAD phenotype and other clinical factors in multivariable analysis. CONCLUSIONS Fulminant fibroproliferation with higher mesenchymal CFU counts in BAL is noted in restrictive allograft syndrome and is independently associated with poor survival after CLAD onset.
Collapse
Affiliation(s)
- Michael P Combs
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Meng Xia
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - David S Wheeler
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth A Belloli
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Natalie M Walker
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Russell R Braeuer
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Dennis M Lyu
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Susan Murray
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Vibha N Lama
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
17
|
Modulation of NLRP3 Inflammasome through Formyl Peptide Receptor 1 (Fpr-1) Pathway as a New Therapeutic Target in Bronchiolitis Obliterans Syndrome. Int J Mol Sci 2020; 21:ijms21062144. [PMID: 32244997 PMCID: PMC7139667 DOI: 10.3390/ijms21062144] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 01/12/2023] Open
Abstract
Chronic rejection is the major leading cause of morbidity and mortality after lung transplantation. Bronchiolitis obliterans syndrome (BOS), a fibroproliferative disorder of the small airways, is the main manifestation of chronic lung allograft rejection. We investigated, using transgenic mice, the mechanisms through which the deficiency of IL-1β/IL-18, Casp-1, or Fpr-1 genes could be protective in an experimental model of BOS, induced in mice by allogeneic heterotopic tracheal transplantation. Fpr-1 KO mice showed a marked reduction in histological markers of BOS and of mast cell numbers compared to other groups. Molecular analyses indicated that the absence of the Fpr-1 gene was able to decrease NF-κB nuclear translocation and modulate NLRP3 inflammasome signaling and the mitogen-activated protein kinase (MAPK) pathway in a more significant way compared to other groups. Additionally, Fpr-1 gene deletion caused a reduction in resistance to the apoptosis, assessed by the TUNEL assay. Immunohistochemical analyses indicated changes in nitrotyrosine, PARP, VEGF, and TGF-β expression associated with the pathology, which were reduced in the absence of the Fpr1 gene more so than by the deletion of IL-1β/IL-18 and Casp-1. We underline the importance of the NLRP3 inflammasome and the pathogenic role of Fpr-1 in experimental models of BOS, which is the result of the modulation of immune cell recruitment together with the modulation of local cellular activation, suggesting this gene as a new target in the control of the pathologic features of BOS.
Collapse
|
18
|
Shukla SD, Swaroop Vanka K, Chavelier A, Shastri MD, Tambuwala MM, Bakshi HA, Pabreja K, Mahmood MQ, O’Toole RF. Chronic respiratory diseases: An introduction and need for novel drug delivery approaches. TARGETING CHRONIC INFLAMMATORY LUNG DISEASES USING ADVANCED DRUG DELIVERY SYSTEMS 2020. [PMCID: PMC7499075 DOI: 10.1016/b978-0-12-820658-4.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Globally, chronic respiratory diseases (CRDs), both communicable and noncommunicable, are among the leading causes of mortality, morbidity, economic and societal burden, and disability-adjusted life years (DALYs). CRDs affect multiple components of respiratory system, including the airways, parenchyma, and pulmonary vasculature. Although noncommunicable respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), interstitial lung disease (ILD), cystic fibrosis (CF), and lung cancer (LC), account for enormous disease burden, the currently available therapies only focus on alleviating the symptoms of diseases rather than providing optimal treatment and/or prevention. Similarly a major respiratory communicable disease, that is, tuberculosis (TB), is associated with the challenge of increasingly developing antibiotic resistance in the bacterial pathogen Mycobacterium tuberculosis. In light of these challenges, we aim to summarize the underlying molecular and cellular mechanisms that lead to hallmark pathophysiology of CRDs. Moreover, we will also highlight the limitations of current therapeutic strategies and explore novel drug delivery options that may be potentially more effective in the management of CRDs.
Collapse
|
19
|
Peripheral CD19+CD24highCD38high B-regulatory cells in lung transplant recipients. Transpl Immunol 2019; 57:101245. [DOI: 10.1016/j.trim.2019.101245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 10/26/2022]
|
20
|
Dong M, Wang X, Guan Y, Li T. MiR-27a-3p downregulation contributes to the development of occlusive bronchiolitis. Cell Stress Chaperones 2019; 24:883-889. [PMID: 31452017 PMCID: PMC6717216 DOI: 10.1007/s12192-019-01026-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 01/06/2023] Open
Abstract
The only effective clinical treatment for many end-stage lung diseases is lung transplantation. However, chronic rejection of transplanted lung affects the long-term efficacy of lung transplantation to a large extent, thereby limiting the clinical application of lung transplantation. Occlusive bronchiolitis (OB) is a major cause of chronic functional loss of the transplanted lung. However, the OB pathogenesis remains unclear. Therefore, studying the OB pathogenesis and finding effective intervention methods are highly important. This study analyzed changes in the expression profile of microRNAs and transcription factors in mice with OB after orthotopic tracheal transplantation. miR-27a-3p was upregulated in lung tissue 20 days after transplantation. Transcription factor microarray analysis revealed that Smad3 was significantly downregulated. A miRNA-mRNA interaction network was constructed, and specific regulatory effects of miR-27a-3p on Smad3 were found. Smad3 was strongly associated with tumorigenesis and organ fibrosis. Compared with the control group, miR-27a-3p inhibited the epithelial-mesenchymal transformation (EMT) of lung epithelial cells. In addition, miR-27a-3p inhibition promoted the invasion and migration of lung epithelial cells. Dual luciferase reporter gene assay showed that miR-27a-3p can regulate the promoter activity of Smad3. MiR-27a-3p also inhibited the expression of inflammatory cytokines. Western blot results showed that miR-27a-3p can upregulate the E-cadherin expression and downregulate the expression of vimentin, fibronectin, and α-SMA. By studying the OB pathogenesis, we found that inhibition or alteration of the occurrence of EMT may reduce the proportion of chronic rejection of lung transplantation. MiR-27a-3p may also be developed as a new drug for the OB therapy. This finding will provide many possibilities for OB treatment and improve the prognosis of patients with OB.
Collapse
Affiliation(s)
- Ming Dong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Anshan Road No. 154, Heping District, Tianjin, 300052, People's Republic of China.
| | - Xin Wang
- Department of Pediatric Surgery, Tianjin Children's Hospital, No. 238 LongYan Road, Tianjin, 300134, People's Republic of China
| | - Yong Guan
- Department of Pediatric Surgery, Tianjin Children's Hospital, No. 238 LongYan Road, Tianjin, 300134, People's Republic of China
| | - Tong Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Anshan Road No. 154, Heping District, Tianjin, 300052, People's Republic of China
| |
Collapse
|
21
|
Ozeki N, Yamawaki-Ogata A, Narita Y, Mii S, Ushida K, Ito M, Hirano SI, Kurokawa R, Ohno K, Usui A. Hydrogen water alleviates obliterative airway disease in mice. Gen Thorac Cardiovasc Surg 2019; 68:158-163. [PMID: 31468277 DOI: 10.1007/s11748-019-01195-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 08/18/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Bronchiolitis obliterans syndrome arising from chronic airway inflammation is a leading cause of death following lung transplantation. Several studies have suggested that inhaled hydrogen can protect lung grafts from ischemia-reperfusion injury via anti-inflammatory and -oxidative mechanisms. We investigated whether molecular hydrogen-saturated water can preserve lung allograft function in a heterotopic tracheal allograft mouse model of obliterative airway disease METHODS: Obliterative airway disease was induced by heterotopically transplanting tracheal allografts from BALB/c donor mice into C57BL/6 recipient mice, which were subsequently administered hydrogen water (10 ppm) or tap water (control group) (n = 6 each) daily without any immunosuppressive treatment. Histological and immunohistochemical analyses were performed on days 7, 14, and 21. RESULTS Hydrogen water decreased airway occlusion on day 14. No significant histological differences were observed on days 7 or 21. The cluster of differentiation 4/cluster of differentiation 3 ratio in tracheal allografts on day 14 was higher in the hydrogen water group than in control mice. Enzyme-linked immunosorbent assay performed on day 7 revealed that hydrogen water reduced the level of the pro-inflammatory cytokine interleukin-6 and increased that of forkhead box P3 transcription factor, suggesting an enhancement of regulatory T cell activity. CONCLUSIONS Hydrogen water suppressed the development of mid-term obliterative airway disease in a mouse tracheal allograft model via anti-oxidant and -inflammatory mechanisms and through the activation of Tregs. Thus, hydrogen water is a potential treatment strategy for BOS that can improve the outcome of lung transplant patients.
Collapse
Affiliation(s)
- Naoki Ozeki
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Aika Yamawaki-Ogata
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Narita
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaori Ushida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Specimen Preparation Room for Optical Microscopic Examinations, Core Clinical Research Hospital Support Room, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiko Usui
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
22
|
Dhillon S, McKinnon E, Wrobel J, Lavender M, Lawrence S, Gabbay E, Musk M. Lung transplant: the Western Australian experience. Intern Med J 2019; 48:1337-1345. [PMID: 29923278 DOI: 10.1111/imj.14001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND The Western Australian lung transplant programme commenced in 2004 to serve the growing demand of patients with end-stage lung disease. AIM This report summarises our 11-year experience in lung transplantation. METHODS Data on 115 consecutive patients and their respective donors transplanted between 2004 and 2015 were collected. The Kaplan-Meier method was used to estimate survival. Cox regression was used to analyse the impact of donor and recipient characteristics on survival. RESULTS A total of 88 bilateral, 22 single-lung and 5 heart-lung transplants were performed in Western Australia during the first 11 years of the lung transplant programme. The most common indications for transplantation were interstitial lung disease (30.4%), cystic fibrosis (27.8%) and chronic obstructive pulmonary disease (excluding alpha-1 antitrypsin deficiency) (22.6%). Median recipient age was 50 years. Overall survival rates were 96% at 3 months, 93% at 1 year, 84% at 3 years and 70% at 5 years. Older age and higher BMI negatively impacted survival. Chronic lung allograft dysfunction was the leading cause of late mortality. CONCLUSION Lung transplantation is a treatment option in end-stage lung disease, with annual transplant rates in Western Australia continuing to rise. Our patients enjoy survival rates that compare favourably against international standards.
Collapse
Affiliation(s)
- Sarbroop Dhillon
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Elizabeth McKinnon
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia, Australia
| | - Jeremy Wrobel
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Melanie Lavender
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Sharon Lawrence
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Eli Gabbay
- The University of Notre Dame, Fremantle, Western Australia, Australia
| | - Michael Musk
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
23
|
Diagnosis, Pathophysiology and Experimental Models of Chronic Lung Allograft Rejection. Transplantation 2019; 102:1459-1466. [PMID: 29683998 DOI: 10.1097/tp.0000000000002250] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chronic rejection is the Achilles heel of modern lung transplantation, characterized by a slow, progressive decline in allograft function. Clinically, this manifests as obstructive disease, restrictive disease, or a mixture of the 2 depending on the underlying pathology. The 2 major phenotypes of chronic rejection include bronchiolitis obliterans syndrome and restrictive allograft syndrome. The last decade of research has revealed that each of these phenotypes has a unique underlying pathophysiology which may require a distinct treatment regimen for optimal control. Insights into the intricate alloimmune pathways contributing to chronic rejection have been gained from both large and small animal models, suggesting directions for future research. In this review, we explore the pathological hallmarks of chronic rejection, recent insights gained from both clinical and basic science research, and the current state of animal models of chronic lung rejection.
Collapse
|
24
|
A Pilot Study to Investigate the Balance between Proteases and α1-Antitrypsin in Bronchoalveolar Lavage Fluid of Lung Transplant Recipients. High Throughput 2019; 8:ht8010005. [PMID: 30781848 PMCID: PMC6480715 DOI: 10.3390/ht8010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
The neutrophilic component in bronchiolitis obliterans syndrome (BOS, the main form of chronic lung rejection), plays a crucial role in the pathogenesis and maintenance of the disorder. Human Neutrophil Elastase (HNE), a serine protease responsible of elastin degradation whose action is counteracted by α1-antitrypsin (AAT), a serum inhibitor specific for this protease. This work aimed to investigate the relationship between HNE and AAT in bronchoalveolar lavage fluid (BALf) from stable lung transplant recipients and BOS patients to understand whether the imbalance between proteases and inhibitors is relevant to the development of BOS. To reach this goal a multidisciplinary procedure was applied which included: (i) the use of electrophoresis/western blotting coupled with liquid chromatography-mass spectrometric analysis; (ii) the functional evaluation of the residual antiprotease activity, and (iii) a neutrophil count. The results of these experiments demonstrated, for the first time, the presence of the complex between HNE and AAT in a number of BALf samples. The lack of this complex in a few specimens analyzed was investigated in relation to a patient’s lung inflammation. The neutrophil count and the determination of HNE and AAT activities allowed us to speculate that the presence of the complex correlated with the level of lung inflammation.
Collapse
|
25
|
A novel mechanism for immune regulation after human lung transplantation. J Thorac Cardiovasc Surg 2019; 157:2096-2106. [PMID: 31288367 PMCID: PMC6625531 DOI: 10.1016/j.jtcvs.2018.12.105] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 11/13/2018] [Accepted: 12/02/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Lung transplantation is therapeutic for end-stage lung disease, but survival is limited due to bronchiolitis obliterans syndrome and restrictive chronic lung allograft dysfunction. We sought a common denominator in lung transplant recipients, analyzing risk factors that trigger immune responses that lead to bronchiolitis obliterans syndrome. METHODS We collected blood from patients who underwent lung transplant at our institution. Exosomes were isolated from the sera of recipients with risk factors for chronic rejection and from stable recipients. Exosomes were analyzed with western blot, using antibodies to lung self-antigens K alpha 1 tubulin and collagen-V, costimulatory molecules (costimulatory molecule 80, costimulatory molecule 86), transcription factors (nuclear factor kappa-light-chain-enhancer of activated B cells, hypoxia-inducible factor 1α, Class II Major Histocompatibility Complex Transactivator), and 20S proteasome. RESULTS Of the 90 patients included, we identified 5 with grade 3 primary graft dysfunction, 5 without, 15 with respiratory viral infection, 10 with acute rejection, 10 with donor-specific antibodies (DSA), 5 without DSA, and 10 who were stable for exosome isolation. Recipients with grade 3 primary graft dysfunction, respiratory viral infection, acute rejection, and DSA had exosomes containing self-antigens; exosomes from stable recipients did not. Exosomes from recipients with grade 3 primary graft dysfunction, acute rejection, and DSA also demonstrated costimulatory molecule 80, costimulatory molecule 86, major histocompatibility complex class II, transcription factor, and 20S proteasome. CONCLUSIONS Transplanted lungs with grade 3 primary graft dysfunction, symptomatic respiratory viral infection, acute rejection, and immune responses induce exosomes that contain self-antigens, costimulatory molecules, major histocompatibility complex class II, transcription factors, and 20S proteasome. Release of circulating exosomes post-transplant from the aforementioned stress-inducing insults augment immunity and may play an important role in the pathogenesis of bronchiolitis obliterans syndrome.
Collapse
|
26
|
Clinical Application of Stem/Stromal Cells in COPD. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121219 DOI: 10.1007/978-3-030-29403-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive life-threatening disease that is significantly increasing in prevalence and is predicted to become the third leading cause of death worldwide by 2030. At present, there are no true curative treatments that can stop the progression of the disease, and new therapeutic strategies are desperately needed. Advances in cell-based therapies provide a platform for the development of new therapeutic approaches in severe lung diseases such as COPD. At present, a lot of focus is on mesenchymal stem (stromal) cell (MSC)-based therapies, mainly due to their immunomodulatory properties. Despite increasing number of preclinical studies demonstrating that systemic MSC administration can prevent or treat experimental COPD and emphysema, clinical studies have not been able to reproduce the preclinical results and to date no efficacy or significantly improved lung function or quality of life has been observed in COPD patients. Importantly, the completed appropriately conducted clinical trials uniformly demonstrate that MSC treatment in COPD patients is well tolerated and no toxicities have been observed. All clinical trials performed so far, have been phase I/II studies, underpowered for the detection of potential efficacy. There are several challenges ahead for this field such as standardized isolation and culture procedures to obtain a cell product with high quality and reproducibility, administration strategies, improvement of methods to measure outcomes, and development of potency assays. Moreover, COPD is a complex pathology with a diverse spectrum of clinical phenotypes, and therefore it is essential to develop methods to select the subpopulation of patients that is most likely to potentially respond to MSC administration. In this chapter, we will discuss the current state of the art of MSC-based cell therapy for COPD and the hurdles that need to be overcome.
Collapse
|
27
|
Takamori S, Shoji F, Okamoto T, Kozuma Y, Matsubara T, Haratake N, Akamine T, Katsura M, Takada K, Toyokawa G, Tagawa T, Maehara Y. HMGB1 blockade significantly improves luminal fibrous obliteration in a murine model of bronchiolitis obliterans syndrome. Transpl Immunol 2018; 53:13-20. [PMID: 30508580 DOI: 10.1016/j.trim.2018.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although high-mobility group box-1 (HMGB1), which is a nuclear protein, was reported to enhance the allogeneic responses in transplantation, the effect of HMGB1 on bronchiolitis obliterans syndrome (BOS) is unknown. METHODS A murine heterotopic tracheal transplantation model was used. Protein concentrations of HMGB1, interferon-γ (IFN-γ), interleukin (IL)-10, and IL-17 were analyzed in the isografts, allografts, controls, and HMGB1-neutralizing antibody administered allografts (n = 6; Days 1, 3, 5, 7, 14, 21, and 28). The luminal fibrous occlusion was analyzed (n = 6; Days 7, 14, 21, and 28). Infiltrating CD8 and CD4 T lymphocytes around the allografts and serum levels of IFN-γ and IL-10 were evaluated (n = 6; Day 7). RESULTS The HMGB1 levels in the allografts were significantly increased compared with the isografts at Day 7. HMGB1 blockade did not change the IL-17 level, but decreased the IFN-γ/IL-10 ratio in the early phase (Days 5 and 7) and significantly improved the fibrous occlusion in the late phase (Days 14, 21, and 28). HMGB1 blockade significantly suppressed the CD8 T lymphocytes infiltration and decreased the serum IFN-γ/IL-10 ratio compared with the control at Day 7. CONCLUSIONS HMGB1 may be a trigger of the BOS pathogenesis and candidate target for the treatment of the disease.
Collapse
Affiliation(s)
- Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Fumihiro Shoji
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan.
| | - Tatsuro Okamoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yuka Kozuma
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Taichi Matsubara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Naoki Haratake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Masakazu Katsura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Kazuki Takada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| |
Collapse
|
28
|
Yang M, Chen G, Zhang X, Ding Z, Miao Y, Yang Y, Chen ZK, Jiang F, Chang S, Zhou P. A novel MyD88 inhibitor attenuates allograft rejection after heterotopic tracheal transplantation in mice. Transpl Immunol 2018; 53:1-6. [PMID: 30472390 DOI: 10.1016/j.trim.2018.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 11/21/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND After lung transplantation, the major complication limiting the long-term survival of allografts is obliterative bronchiolitis (OB), characterized by chronic rejection. Innate immune responses contribute to the development of OB. In this study, we used a murine heterotopic tracheal transplantation mouse model to examine the effects of a newtype of innate immune inhibitor, TJ-M2010-5. METHODS Syngeneic tracheal grafts were transplanted heterotopically from C57BL/6 mice to C57BL/6 mice. Allografts from BALB/c mice were transplanted to C57BL/6 mice. The allograft recipients were treated with TJ-M2010-5, and anti-mouse CD154 (MR-1). The grafts were harvested at 7, 14, and 28 days and evaluated by histological and real-time RT-PCR analyses. RESULTS In untreated allografts, almost all epithelial cells fell off at 7 days and tracheal occlusion reached a peak at 28 days. However, the loss of the epithelium and airway obstruction were significantly improved in mice treated with TJ-M2010-5 combined with MR-1. The relative mRNA expression levels of pro-inflammatory cytokines were upregulated in allogeneic tracheal grafts, and treatment with the two drugs reduced the production of pro-inflammatory cytokines and infiltration of inflammatory cells. CONCLUSIONS In heterotopic tracheal transplantation models, TJ-M2010-5 combined with MR-1 could ameliorate the development of OB.
Collapse
Affiliation(s)
- Min Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Gen Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zuochuan Ding
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Yan Miao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Yang Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Zhonghua Klaus Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China
| | - Fengchao Jiang
- Academy of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China.
| | - Ping Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 43003, China.
| |
Collapse
|
29
|
Vella S, Conaldi PG, Cova E, Meloni F, Liotta R, Cuzzocrea S, Martino L, Bertani A, Luca A, Vitulo P. Lung resident mesenchymal cells isolated from patients with the Bronchiolitis Obliterans Syndrome display a deregulated epigenetic profile. Sci Rep 2018; 8:11167. [PMID: 30042393 PMCID: PMC6057887 DOI: 10.1038/s41598-018-29504-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022] Open
Abstract
Bronchiolitis Obliterans Syndrome is the major determinant of the graft function loss after lung transplantation, but its pathogenesis is still incompletely understood and currently available therapeutic strategies are poorly effective. A deeper understanding of its pathogenic mechanisms is crucial for the development of new strategies to prevent and treat this devastating complication. In this study, we focused on the mesenchymal stromal cells, recently recognized as BOS key effectors, and our primary aim was to identify their epigenetic determinants, such as histone modifications and non-coding RNA regulation, which could contribute to their differentiation in myofibroblasts. Interestingly, we identified a deregulated expression of histone deacetylases and methyltransferases, and a microRNA-epigenetic regulatory network, which could represent novel targets for anti-fibrotic therapy. We validated our results in vitro, in a cell model of fibrogenesis, confirming the epigenetic involvement in this process and paving the way for a new application for epigenetic drugs.
Collapse
Affiliation(s)
- Serena Vella
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy.
- Anemocyte S.r.l, Gerenzano, Italy.
| | - Pier Giulio Conaldi
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Emanuela Cova
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Federica Meloni
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Rosa Liotta
- Department of Diagnostic and Therapeutic Services, Pathology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Lavinia Martino
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Alessandro Bertani
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Angelo Luca
- Department of Diagnostic and Therapeutic Services, Radiology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Patrizio Vitulo
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| |
Collapse
|
30
|
Pecoraro Y, Carillo C, Diso D, Mantovani S, Cimino G, De Giacomo T, Troiani P, Shafii M, Gherzi L, Amore D, Rendina EA, Venuta F, Anile M. Efficacy of Extracorporeal Photopheresis in Patients With Bronchiolitis Obliterans Syndrome After Lung Transplantation. Transplant Proc 2017; 49:695-698. [PMID: 28457374 DOI: 10.1016/j.transproceed.2017.02.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Lung transplantation (LT) is only therapeutic option for patients affected by chronic respiratory failure. Chronic rejection, also known as bronchiolitis obliterans syndrome (BOS), is still the main cause of death and the most important factor that influences post-transplantation quality of life. Currently available therapies have not been proven to result in significant benefit in the prevention or treatment of BOS. Extracorporeal photopheresis (ECP) seems to reduce the rate of lung function decline in transplant recipients with progressive BOS. METHODS From 1991 until now, 239 LTs were performed at our center. Fifty-four patients (22.5%) developed BOS; 15 of these (27.7%) were treated with ECP. At the beginning of the treatment, all patients showed a mean decline of forced expiratory volume in 1 second (FEV1) from baseline values of 45.8% ± 17.2%; 2 patients were in long-term oxygen therapy. RESULTS Mean follow-up from November 2013 to June 2016 was 11.6 ± 7 months. Twelve patients (80%) showed lung function stabilization with an FEV1 range after treatment between -6% to +8% from the pre-treatment values. We did not report any adverse effects or increase of infections incidence. DISCUSSION ECP seems to be an effective and well-tolerated therapeutic option for LT patients with BOS in terms of stabilization of lung function and increased survival.
Collapse
Affiliation(s)
- Y Pecoraro
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy.
| | - C Carillo
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - D Diso
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - S Mantovani
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - G Cimino
- Department of Pediatrics, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - T De Giacomo
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - P Troiani
- Department of Pediatrics, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - M Shafii
- Department of Hematology, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - L Gherzi
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - D Amore
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - E A Rendina
- Department of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - F Venuta
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| | - M Anile
- Department of Thoracic Surgery, Policlinico Umberto I, Sapienza University of Rome, Italy
| |
Collapse
|
31
|
Role of Circulating MicroRNAs in the Immunopathogenesis of Rejection After Pediatric Lung Transplantation. Transplantation 2017; 101:2461-2468. [PMID: 27941431 DOI: 10.1097/tp.0000000000001595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Acute rejection (AR) and development of chronic rejection, bronchiolitis obliterans syndrome (BOS) remain major limiting factors for lung transplantation (LTx). This retrospective study is to identify differentially expressed circulating microRNAs (miRNAs) that associate with development of AR and BOS in pediatric lung transplant recipients (LTxR). METHODS We determined the circulating levels of 7 selected candidate miRNAs in 14 LTxR with AR, 7 with BOS, and compared them against 13 stable pediatric LTxR at 1, 6, and 12 months after LTx. In addition, 6 AR, 7 BOS, and 8 stable pediatric LTxR, 16 AR, 17 BOS, and 16 stable adult LTxR were included for validation. RESULTS MiR-10a, -195, -133b were significantly lower in AR and miR-144, -142-5p, -155 were higher in AR compared to stable (P < 0.05). In addition, circulating levels of miR-134, -10a, -195, -133b were significantly lower and miR-144, -142-5p, -155 were higher (P < 0.05) with development of BOS. The receiver-operating characteristic demonstrated that miR-142-5p, miR-155, and miR-195 strongly discriminated patients with AR from stable LTxR (P < 0.001 for all comparisons): miR-142-5p (area under the curve [AUC], 0.854), miR-155 (AUC, 0.876), and miR-195 (AUC, 0.872). Further, miR-10a, miR-142-5p, miR-144, and miR-155 strongly discriminated BOS from stable LTxR (P < 0.001 for all comparisons). CONCLUSIONS We demonstrated that differential expression of circulating miRNAs occurs in LTxR with AR and BOS, suggesting that they can provide not only important clues to pathogenesis but also may serve as potential noninvasive biomarkers for AR and BOS after pediatric LTx.
Collapse
|
32
|
The Presence of Pretransplant HLA Antibodies Does Not Impact the Development of Chronic Lung Allograft Dysfunction or CLAD-Related Death. Transplantation 2017; 101:2207-2212. [DOI: 10.1097/tp.0000000000001494] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Benden C, Haughton M, Leonard S, Huber LC. Therapy options for chronic lung allograft dysfunction–bronchiolitis obliterans syndrome following first-line immunosuppressive strategies: A systematic review. J Heart Lung Transplant 2017; 36:921-933. [DOI: 10.1016/j.healun.2017.05.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 11/27/2022] Open
|
34
|
Lee S, Yamauchi T, Ishii N, Hashimoto T, Kinoshita K, Imamura S, Kamiya K. Achievement of the longest survival of paraneoplastic pemphigus with bronchiolitis obliterans associated with follicular lymphoma using R-CHOP chemotherapy. Int J Hematol 2017; 106:852-859. [PMID: 28791608 DOI: 10.1007/s12185-017-2305-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 11/30/2022]
Abstract
Paraneoplastic pemphigus (PNP) is a rare, fatal, paraneoplastic autoimmune mucocutaneous blistering disease, commonly associated with lymphoproliferative disorders, including malignant lymphomas. Lymphoproliferative disorders associated with PNP are sometimes associated with a serious lung complication, bronchiolitis obliterans (BO). Due to its rarity, guidelines for the management of PNP have not been established. Furthermore, most patients die within 1 year. Here we report the successful treatment of lymphoma-associated PNP and BO using R-CHOP chemotherapy. A 53-year-old Japanese man was admitted to our hospital for severe erosive stomatitis. Computed tomography and positron emission tomography showed multiple lymphadenopathies. He was diagnosed with follicular lymphoma (Ann Arbor stage IVA) and PNP-related BO. The patient underwent six cycles of R-CHOP and an additional cycle of rituximab. Both the erosive stomatitis and the obstructive lung disease persisted, but complete response of the follicular lymphoma was achieved. The patient survived 27 months after diagnosis. Although he died from progressive respiratory failure due to BO, we note that this patient achieved the longest survival of any reported case of PNP-related BO associated with a lymphoproliferative disorder. The present case suggests that intensive immunochemotherapy for underlying lymphoma may improve the prognosis in patients with PNP-related BO associated with lymphoma.
Collapse
Affiliation(s)
- Shin Lee
- Department of Hematology, Japan Red Cross Fukui Hospital, 2-4-1 Tsukimi, Fukui, Fukui, 918-8501, Japan.
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Norito Ishii
- Department of Dermatology, School of Medicine, Kurume University, Fukuoka, Japan
| | - Takashi Hashimoto
- Kurume University Institute of Cutaneous Cell Biology, Fukuoka, Japan
| | - Keiichi Kinoshita
- Department of Hematology, Japan Red Cross Fukui Hospital, 2-4-1 Tsukimi, Fukui, Fukui, 918-8501, Japan
| | - Shin Imamura
- Department of Hematology, Japan Red Cross Fukui Hospital, 2-4-1 Tsukimi, Fukui, Fukui, 918-8501, Japan
| | - Kenichi Kamiya
- Department of Hematology, Japan Red Cross Fukui Hospital, 2-4-1 Tsukimi, Fukui, Fukui, 918-8501, Japan
| |
Collapse
|
35
|
Analysis of long term CD4+CD25highCD127- T-reg cells kinetics in peripheral blood of lung transplant recipients. BMC Pulm Med 2017; 17:102. [PMID: 28720146 PMCID: PMC5516333 DOI: 10.1186/s12890-017-0446-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Background The role of CD4+CD25highCD127− T-reg cells in solid-organ Transplant (Tx) acceptance has been extensively studied. In previous studies on kidney and liver recipients, peripheral T-reg cell counts were associated to graft survival, while in lung Tx, there is limited evidence for similar findings. This study aims to analyze long term peripheral kinetics of T-reg-cells in a cohort of lung recipients and tests its association to several clinical variables. Methods From jan 2009 to dec 2014, 137 lung Tx recipients were submitted to an immunological follow up (median: 105.9 months (6.7–310.5)). Immunological follow up consisted of a complete blood peripheral immuno-phenotype, inclusive of CD4+CD25highCD127− T and FOXP3+ cells. We tested the association between T-reg and relevant variables by linear OR regression models for repeated measures, adjusting for time from Tx. Also, by ordered logistic models for panel data, the association between Chronic Lung Allograft Dysfuncton (CLAD) onset/progression and T-reg counts in the previous 3 months was tested. Results Among all variables analyzed at multivariate analysis: Bronchiolitis Obliterans Syndrome (OR −6.51, p < 0.001), Restrictive Allograft Syndrome (OR −5.19, p = 0.04) and Extracorporeal photopheresis (OR −5.65, p < 0.001) were significantly associated to T-reg cell. T-reg cell counts progressively decreased according to the severity of CLAD. Furthermore, patients with higher mean T-reg counts in a trimester had a significantly lower risk (OR 0.97, p = 0.012) of presenting CLAD or progressing in the graft dysfunction in the following trimester. Conclusions Our present data confirm animal observations on the possible role of T-reg in the evolution of CLAD.
Collapse
|
36
|
Microbiome in the pathogenesis of cystic fibrosis and lung transplant-related disease. Transl Res 2017; 179:84-96. [PMID: 27559681 DOI: 10.1016/j.trsl.2016.07.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 01/01/2023]
Abstract
Significant advances in culture-independent methods have expanded our knowledge about the diversity of the lung microbial environment. Complex microorganisms and microbial communities can now be identified in the distal airways in a variety of respiratory diseases, including cystic fibrosis (CF) and the posttransplantation lung. Although there are significant methodologic concerns about sampling the lung microbiome, several studies have now shown that the microbiome of the lower respiratory tract is distinct from the upper airway. CF is a disease characterized by chronic airway infections that lead to significant morbidity and mortality. Traditional culture-dependent methods have identified a select group of pathogens that cause exacerbations in CF, but studies using bacterial 16S rRNA gene-based microarrays have shown that the CF microbiome is an intricate and dynamic bacterial ecosystem, which influences both host immune health and disease pathogenesis. These microbial communities can shift with external influences, including antibiotic exposure. In addition, there have been a number of studies suggesting a link between the gut microbiome and respiratory health in CF. Compared with CF, there is significantly less knowledge about the microbiome of the transplanted lung. Risk factors for bronchiolitis obliterans syndrome, one of the leading causes of death, include microbial infections. Lung transplant patients have a unique lung microbiome that is different than the pretransplanted microbiome and changes with time. Understanding the host-pathogen interactions in these diseases may suggest targeted therapies and improve long-term survival in these patients.
Collapse
|
37
|
Lung Transplant Rejection and Surveillance in 2016: Newer Options. CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Identification of miRNAs Potentially Involved in Bronchiolitis Obliterans Syndrome: A Computational Study. PLoS One 2016; 11:e0161771. [PMID: 27564214 PMCID: PMC5001701 DOI: 10.1371/journal.pone.0161771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of Bronchiolitis Obliterans Syndrome (BOS), the main clinical phenotype of chronic lung allograft dysfunction, is poorly understood. Recent studies suggest that epigenetic regulation of microRNAs might play a role in its development. In this paper we present the application of a complex computational pipeline to perform enrichment analysis of miRNAs in pathways applied to the study of BOS. The analysis considered the full set of miRNAs annotated in miRBase (version 21), and applied a sequence of filtering approaches and statistical analyses to reduce this set and to score the candidate miRNAs according to their potential involvement in BOS development. Dysregulation of two of the selected candidate miRNAs–miR-34a and miR-21 –was clearly shown in in-situ hybridization (ISH) on five explanted human BOS lungs and on a rat model of acute and chronic lung rejection, thus definitely identifying miR-34a and miR-21 as pathogenic factors in BOS and confirming the effectiveness of the computational pipeline.
Collapse
|
39
|
Low Levels of Exhaled Surfactant Protein A Associated With BOS After Lung Transplantation. Transplant Direct 2016; 2:e103. [PMID: 27795995 PMCID: PMC5068199 DOI: 10.1097/txd.0000000000000615] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/08/2016] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND There is no clinically available marker for early detection or monitoring of chronic rejection in the form of bronchiolitis obliterans syndrome (BOS), the main long-term complication after lung transplantation. Sampling and analysis of particles in exhaled air is a valid, noninvasive method for monitoring surfactant protein A (SP-A) and albumin in the distal airways. METHODS We asked whether differences in composition of exhaled particles can be detected when comparing stable lung transplant recipients (LTRs) (n = 26) with LTRs who develop BOS (n = 7). A comparison between LTRs and a matching group of healthy controls (n = 33) was also conducted. Using a system developed in-house, particles were collected from exhaled air by the principal of inertial impaction before chemical analysis by immunoassays. RESULTS Surfactant protein A in exhaled particles and the SP-A/albumin ratio were lower (P = 0.002 and P = 0.0001 respectively) in the BOS group compared to the BOS-free group. LTRs exhaled higher amount of particles (P < 0.0001) and had lower albumin content (P < 0.0001) than healthy controls. CONCLUSIONS We conclude that low levels of SP-A in exhaled particles are associated with increased risk of BOS in LTRs. The possibility that this noninvasive method can be used to predict BOS onset deserves further study with prospective and longitudinal approaches.
Collapse
|
40
|
Rolandsson Enes S, Andersson Sjöland A, Skog I, Hansson L, Larsson H, Le Blanc K, Eriksson L, Bjermer L, Scheding S, Westergren-Thorsson G. MSC from fetal and adult lungs possess lung-specific properties compared to bone marrow-derived MSC. Sci Rep 2016; 6:29160. [PMID: 27381039 PMCID: PMC4933903 DOI: 10.1038/srep29160] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 06/16/2016] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are multipotent cells with regenerative and immune-modulatory properties. Therefore, MSC have been proposed as a potential cell-therapy for bronchiolitis obliterans syndrome (BOS). On the other hand, there are publications demonstrating that MSC might be involved in the development of BOS. Despite limited knowledge regarding the functional role of tissue-resident lung-MSC, several clinical trials have been performed using MSC, particularly bone marrow (BM)-derived MSC, for various lung diseases. We aimed to compare lung-MSC with the well-characterized BM-MSC. Furthermore, MSC isolated from lung-transplanted patients with BOS were compared to patients without BOS. Our study show that lung-MSCs are smaller, possess a higher colony-forming capacity and have a different cytokine profile compared to BM-MSC. Utilizing gene expression profiling, 89 genes including lung-specific FOXF1 and HOXB5 were found to be significantly different between BM-MSC and lung-MSC. No significant differences in cytokine secretion or gene expression were found between MSC isolated from BOS patients compared recipients without BOS. These data demonstrate that lung-resident MSC possess lung-specific properties. Furthermore, these results show that MSC isolated from lung-transplanted patients with BOS do not have an altered phenotype compared to MSC isolated from good outcome recipients.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| | | | - Ingrid Skog
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Lennart Hansson
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Hillevi Larsson
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Katarina Le Blanc
- Division of Clinical Immunology; Centre for Allogeneic Stem Cell Transplantation, Karolinska Institutet, Huddinge University Hospital, Stockholm, Sweden
| | - Leif Eriksson
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Stefan Scheding
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Hematology, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
41
|
Diagnostic value of plasma and bronchoalveolar lavage samples in acute lung allograft rejection: differential cytology. Respir Res 2016; 17:74. [PMID: 27323950 PMCID: PMC4915079 DOI: 10.1186/s12931-016-0391-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022] Open
Abstract
Diagnosis of acute lung allograft rejection is currently based on transbronchial lung biopsies. Additional methods to detect acute allograft dysfunction derived from plasma and bronchoalveolar lavage samples might facilitate diagnosis and ultimately improve allograft survival. This review article gives an overview of the cell profiles of bronchoalveolar lavage and plasma samples during acute lung allograft rejection. The value of these cells and changes within the pattern of differential cytology to support the diagnosis of acute lung allograft rejection is discussed. Current findings on the topic are highlighted and trends for future research are identified.
Collapse
|
42
|
Chen QR, Wang LF, Xia SS, Zhang YM, Xu JN, Li H, Ding YZ. Role of interleukin-17A in early graft rejection after orthotopic lung transplantation in mice. J Thorac Dis 2016; 8:1069-79. [PMID: 27293822 DOI: 10.21037/jtd.2015.12.08] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The cellular and molecular mechanisms underlying lung allograft rejection remain poorly understood. We investigated the potential role of interleukin (IL)-17A in lung transplant rejection in a mouse model, because previous studies in clinical and rodent models have implicated IL-17A in both acute and chronic rejection. METHODS To generate an orthotopic lung transplantation model, lungs from C57BL/6 or BALB/c mice were transplanted into C57BL/6 mice (isograft and allograft models, respectively). The effects of anti-IL-17A treatment in allograft recipients were investigated. The histological features and rejection status of isografts and allografts were assessed at 3, 7, and 28 days after transplantation, and differences in graft infiltrating cells and mRNA expression of relevant cytokines were quantified at 3 and 7 days after transplantation. RESULTS As expected, isografts showed no obvious signs of rejection, whereas allografts exhibited minimal-to-mild rejection (grade A1-A2) by day 3 and moderate-to-severe rejection (grade A3-A4) by day 7, without evidence of obliterative bronchiolitis (OB). However, by 28 days, evidence of OB was observed in 67% (2/3) of allografts and severe rejection (grade A4) was observed in all. IL-17 mRNA expression in allografts was increased with rejection, and interferon (IFN)-γ and IL-6 mRNA expression levels followed a similar pattern. In contrast, IL-22 expression in allografts was only slightly increased. Antibody (Ab) neutralization of IL-17A diminished the signs of acute rejection at 7 days after transplantation in allografts, and this early protection was accompanied by a decrease in cellular stress according to histological evaluation, suggesting the involvement of IL-17A in the development of early post-transplantation lesions. CONCLUSIONS Our data indicate that IL-17A is important in the pathophysiology of allograft rejection, and neutralization of IL-17A is a potential therapeutic strategy to preventing lung transplant rejection.
Collapse
Affiliation(s)
- Qi-Rui Chen
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Li-Feng Wang
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Si-Si Xia
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Ya-Mei Zhang
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Jiang-Nan Xu
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Hui Li
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Yao-Zhong Ding
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| |
Collapse
|
43
|
Borthwick LA, Suwara MI, Carnell SC, Green NJ, Mahida R, Dixon D, Gillespie CS, Cartwright TN, Horabin J, Walker A, Olin E, Rangar M, Gardner A, Mann J, Corris PA, Mann DA, Fisher AJ. Pseudomonas aeruginosa Induced Airway Epithelial Injury Drives Fibroblast Activation: A Mechanism in Chronic Lung Allograft Dysfunction. Am J Transplant 2016; 16:1751-65. [PMID: 26714197 PMCID: PMC4879508 DOI: 10.1111/ajt.13690] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 12/03/2015] [Accepted: 12/06/2015] [Indexed: 01/25/2023]
Abstract
Bacterial infections after lung transplantation cause airway epithelial injury and are associated with an increased risk of developing bronchiolitis obliterans syndrome. The damaged epithelium is a source of alarmins that activate the innate immune system, yet their ability to activate fibroblasts in the development of bronchiolitis obliterans syndrome has not been evaluated. Two epithelial alarmins were measured longitudinally in bronchoalveolar lavages from lung transplant recipients who developed bronchiolitis obliterans syndrome and were compared to stable controls. In addition, conditioned media from human airway epithelial cells infected with Pseudomonas aeruginosa was applied to lung fibroblasts and inflammatory responses were determined. Interleukin-1 alpha (IL-1α) was increased in bronchoalveolar lavage of lung transplant recipients growing P. aeruginosa (11.5 [5.4-21.8] vs. 2.8 [0.9-9.4] pg/mL, p < 0.01) and was significantly elevated within 3 months of developing bronchiolitis obliterans syndrome (8.3 [1.4-25.1] vs. 3.6 [0.6-17.1] pg/mL, p < 0.01), whereas high mobility group protein B1 remained unchanged. IL-1α positively correlated with elevated bronchoalveolar lavage IL-8 levels (r(2) = 0.6095, p < 0.0001) and neutrophil percentage (r(2) = 0.25, p = 0.01). Conditioned media from P. aeruginosa infected epithelial cells induced a potent pro-inflammatory phenotype in fibroblasts via an IL-1α/IL-1R-dependent signaling pathway. In conclusion, we propose that IL-1α may be a novel therapeutic target to limit Pseudomonas associated allograft injury after lung transplantation.
Collapse
Affiliation(s)
- L. A. Borthwick
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - M. I. Suwara
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - S. C. Carnell
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - N. J. Green
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - R. Mahida
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - D. Dixon
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - C. S. Gillespie
- School of Mathematics and StatisticsNewcastle UniversityNewcastle upon TyneUK
| | - T. N. Cartwright
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - J. Horabin
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - A. Walker
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - E. Olin
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - M. Rangar
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK,Institute of TransplantationNewcastle Upon Tyne Hospitals NHS Foundation TrustFreeman HospitalNewcastle upon TyneUK
| | - A. Gardner
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - J. Mann
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - P. A. Corris
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK,Institute of TransplantationNewcastle Upon Tyne Hospitals NHS Foundation TrustFreeman HospitalNewcastle upon TyneUK
| | - D. A. Mann
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - A. J. Fisher
- Tissue Fibrosis and Repair GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK,Institute of TransplantationNewcastle Upon Tyne Hospitals NHS Foundation TrustFreeman HospitalNewcastle upon TyneUK
| |
Collapse
|
44
|
Piloni D, Magni S, Oggionni T, Benazzo A, Stella G, Scudeller L, Morosini M, Cova E, Meloni F. Clinical utility of CD4+ function assessment (ViraCor-IBT ImmuKnow test) in lung recipients. Transpl Immunol 2016; 37:35-39. [PMID: 27095000 DOI: 10.1016/j.trim.2016.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 11/19/2022]
Abstract
The ImmuKnow assay measures cell-mediated immunity, quantifying ATP production from peripheral blood CD4+T-cells in solid-organ transplant patients who undergo immunosuppressive therapy. We aimed to measure functional immunity in lung transplant recipients and correlate Immuknow values with immunosuppression levels, presence of chronic lung allograft dysfunction (CLAD) and infections. We evaluated 61 lung recipients who underwent follow-up for lung transplantation between 2010 and 2014. Rejection and infection were retrospectively analyzed. The association between over-immunosuppression and a number of predictors was assessed by means of univariate and multivariate logistic regression models. 71 out of 127 samples (56%) showed an over-immunosuppression with an ImmuKnow assay mean level of 112.92ng/ml (SD±58.2), vs. 406.14ng/ml (SD±167.7) of the rest of our cohort. In the over-immunosuppression group we found 51 episodes of infection (71%) (OR 2.754, 95% CI 1.40-5.39; P-value 0.003). In the other group, only 25 samples (44%) were taken during an infectious episode. The mean absolute ATP level was significantly different between patients with or without infection (202.38±139.06ng/ml vs. 315.51±221.60ng/ml; P<0.001). RAS (Restrictive allograft syndrome) was associated to low ImmuKnow level (P<0.001). These results were confirmed by the multivariate analysis. The ImmuKnow assay levels were significantly lower in infected lung transplant recipients compared with non-infected recipients and in RAS patients.
Collapse
Affiliation(s)
- Davide Piloni
- Department of Internal Medicine, PhD in Experimental Medicine, University of Pavia, Pavia, Italy.
| | - Sara Magni
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Tiberio Oggionni
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alberto Benazzo
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Giulia Stella
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Luigia Scudeller
- Clinical Epidemiology and Biometric Unit, Scientific Direction, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Monica Morosini
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Emanuela Cova
- Cardiothoracic and Vascular Department, Pneumology Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Federica Meloni
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
45
|
Xu J, Chen G, Song T, Zhu Z, Feng Y, Chang H, Chen F, Ma X, Wu D. [Study on the correlation between CMV reactivation and bronchiolitis obliteans after allogeneic hematopoietic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 36:389-92. [PMID: 26031524 PMCID: PMC7342589 DOI: 10.3760/cma.j.issn.0253-2727.2015.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
目的 探讨异基因造血干细胞移植(allo-HSCT)术后CMV重激活与闭塞性细支气管炎(BO)的相互关系。 方法 2011年1月至2013年12月行allo-HSCT的769例患者,通过荧光定量PCR方法检测CMV-DNA水平,免疫荧光染色白细胞PP65抗原血症两种检测方法诊断CMV感染,将CMV感染患者出现BO与未发生CMV感染患者出现BO进行比较,分析CMV感染与BO相关性,并将符合CMV感染患者中出现BO与未出现BO者进行对照分析。 结果 诊断CMV感染259例患者中出现BO 32例(12.35%),而未感染CMV的510例患者发生BO 8例(1.56%),二者差异有统计学意义(P< 0.001)。将诊断CMV感染259例患者,发生BO 32例与未发生BO 227例的二组临床资料进行CMV相关临床分析比较。CMV病毒载量在105拷贝数/ml较低病毒载量102拷贝数/ml出现BO差异有统计学意义。 结论 在allo-HSCT后导致BO的风险因素中,CMV感染是值得注意的相关因素之一,其中高病毒载量和CMV重激活和CMV肺炎是可能危险因素。
Collapse
Affiliation(s)
- Jing Xu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Guanghua Chen
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tiemei Song
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ziling Zhu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yufeng Feng
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Huirong Chang
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Feng Chen
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiao Ma
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Depei Wu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
46
|
Matsuda Y, Wang X, Oishi H, Guan Z, Saito M, Liu M, Keshavjee S, Chow CW. Spleen Tyrosine Kinase Modulates Fibrous Airway Obliteration and Associated Lymphoid Neogenesis After Transplantation. Am J Transplant 2016; 16:342-52. [PMID: 26308240 DOI: 10.1111/ajt.13442] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 06/16/2015] [Accepted: 06/21/2015] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction, the major cause of death following lung transplantation, usually manifests as irreversible airflow obstruction associated with obliterative bronchiolitis (OB), a lesion characterized by chronic inflammation, lymphoid neogenesis, fibroproliferation and small airway obliteration. Spleen tyrosine kinase (Syk), a tyrosine kinase that regulates B cell function and innate immunity, has been implicated in the pathogenesis of chronic inflammation and tissue repair. This study evaluated the role of Syk in development of OB, using an intrapulmonary tracheal transplant model of OB with the conditional Syk-knockout Syk(flox/flox) //rosa26-CreER(T2) mice and a Syk-selective inhibitor, GSK2230413. BALB/c trachea allografts were transplanted into Syk-knockout (Syk(del/del) ) mice or wild-type C57BL/6 recipients treated with GSK2230413. At day 28, histological analysis revealed that in the Syk(del/del) and GSK2230413-treated C57BL/6 recipients, the graft lumen remained open compared with allografts transplanted into Syk-expressing (Syk(flox/flox) ) and placebo control-treated C57BL/6 recipients. Immunofluorescence showed lymphoid neogenesis with distinct B and T cell zones in control mice. In contrast, lymphoid neogenesis was absent and few B or T cells were found in Syk(del/del) and GSK2230413-treated mice. These observations suggest that inhibition of Syk may be a potential therapeutic strategy for the management of OB following lung transplantation.
Collapse
Affiliation(s)
- Y Matsuda
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - X Wang
- Division of Respirology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - H Oishi
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Z Guan
- Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - M Saito
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - M Liu
- Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - S Keshavjee
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Laboratories, Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - C-W Chow
- The Toronto Lung Transplant Program, University of Toronto, Toronto, Canada.,Division of Respirology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
47
|
Jonigk D, Izykowski N, Rische J, Braubach P, Kühnel M, Warnecke G, Lippmann T, Kreipe H, Haverich A, Welte T, Gottlieb J, Laenger F. Molecular Profiling in Lung Biopsies of Human Pulmonary Allografts to Predict Chronic Lung Allograft Dysfunction. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3178-88. [PMID: 26476349 DOI: 10.1016/j.ajpath.2015.08.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/03/2015] [Accepted: 08/28/2015] [Indexed: 10/22/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is the main reason for poor long-term outcome of lung transplantation, with bronchiolitis obliterans (BO) representing the predominant pathological feature. BO is defined as a progressive fibrous obliteration of the small airways, thought to be triggered by a combination of nonimmune bronchial injury and alloimmune and autoimmune mechanisms. Because biopsy samples are too insensitive to reliably detect BO and a decline in lung function test results, which is clinically used to define CLAD, does not detect early stages, there is need for alternative biomarkers for early diagnosis. Herein, we analyzed the cellular composition and differential expression of 45 tissue remodeling-associated genes in transbronchial lung biopsy specimens from two cohorts with 18 patients each: patients who did not develop CLAD within 3 years after transplantation (48 biopsy specimens) and patients rapidly developing CLAD within the first 3 postoperative years (57 biopsy specimens). Integrating the mRNA expression levels of the five most significantly dysregulated genes from the transforming growth factor-β axis (BMP4, IL6, MMP1, SMAD1, and THBS1) into a score, patient groups could be confidently separated and the outcome predicted (P < 0.001). We conclude that overexpression of fibrosis-associated genes may be valuable as a tissue-based molecular biomarker to more accurately diagnose or predict the development of CLAD.
Collapse
Affiliation(s)
- Danny Jonigk
- Institute of Pathology, Hanover Medical School, Hanover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany.
| | - Nicole Izykowski
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Johanna Rische
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Peter Braubach
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Mark Kühnel
- Institute of Functional and Applied Anatomy, Hanover Medical School, Hanover, Germany
| | - Gregor Warnecke
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Thoracic Surgery, Hanover Medical School, Hanover, Germany
| | - Torsten Lippmann
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Hans Kreipe
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Axel Haverich
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Thoracic Surgery, Hanover Medical School, Hanover, Germany
| | - Tobias Welte
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
| | - Jens Gottlieb
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
| | - Florian Laenger
- Institute of Pathology, Hanover Medical School, Hanover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany
| |
Collapse
|
48
|
Belloli EA, Wang X, Murray S, Forrester G, Weyhing A, Lin J, Ojo T, Lama VN. Longitudinal Forced Vital Capacity Monitoring as a Prognostic Adjunct after Lung Transplantation. Am J Respir Crit Care Med 2015; 192:209-18. [PMID: 25922973 DOI: 10.1164/rccm.201501-0174oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE After lung transplantation, spirometric values are routinely followed to assess graft function. FEV1 is used to characterize chronic allograft dysfunction, whereas the course of FVC change has been less acknowledged and rarely used. OBJECTIVES To better understand the temporal relationship and prognostic ability of FEV1 and FVC decline after lung transplantation. METHODS Serial FEV1 and FVC values were studied among 205 bilateral lung transplant recipients. Different decline patterns were characterized and evaluated for prognostic value via restricted mean modeling of mortality and times to other pertinent events. MEASUREMENTS AND MAIN RESULTS Baseline FEV1 was achieved earlier than baseline FVC (median, 296 vs. 378 d; P < 0.0001). Decline in FEV1 or FVC from their respective post-transplant baselines occurred in 85 patients (41%). Fifty-nine of 85 (69%) had an isolated FEV1 decline, with 80% later meeting the FVC decline criterion. This subsequent FVC decline was associated with worsening FEV1 and lower median survival. Twenty-five of 85 patients (29%) demonstrated concurrent FEV1 and FVC decline. Patients with concurrent decline had higher 1- and 5-year mortality rates (1-yr, 53% vs. 18%, P < 0.0001; 5-yr, 61% vs. 48%, P = 0.001). These patients were more likely to have rapid-onset of spirometry decline (P = 0.05) and lower FEV1% predicted (P = 0.04) at presentation. CONCLUSIONS FVC decline from its post-transplant baseline provides valuable prognostic information. Concurrent FEV1 and FVC decline identifies patients with fulminant, rapid deterioration and is the strongest clinical predictor of poor survival. Subsequent FVC decline in patients with an initial isolated FEV1 decline identifies disease progression and portends poor prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Jules Lin
- 4 Division of Thoracic Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Tammy Ojo
- 1 Division of Pulmonary and Critical Care Medicine
| | - Vibha N Lama
- 1 Division of Pulmonary and Critical Care Medicine
| |
Collapse
|
49
|
Afonso Júnior JE, Werebe EDC, Carraro RM, Teixeira RHDOB, Fernandes LM, Abdalla LG, Samano MN, Pêgo-Fernandes PM. Lung transplantation. EINSTEIN-SAO PAULO 2015; 13:297-304. [PMID: 26154550 PMCID: PMC4943827 DOI: 10.1590/s1679-45082015rw3156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 02/08/2015] [Indexed: 11/21/2022] Open
Abstract
Lung transplantation is a globally accepted treatment for some advanced lung diseases, giving the recipients longer survival and better quality of life. Since the first transplant successfully performed in 1983, more than 40 thousand transplants have been performed worldwide. Of these, about seven hundred were in Brazil. However, survival of the transplant is less than desired, with a high mortality rate related to primary graft dysfunction, infection, and chronic graft dysfunction, particularly in the form of bronchiolitis obliterans syndrome. New technologies have been developed to improve the various stages of lung transplant. To increase the supply of lungs, ex vivo lung reconditioning has been used in some countries, including Brazil. For advanced life support in the perioperative period, extracorporeal membrane oxygenation and hemodynamic support equipment have been used as a bridge to transplant in critically ill patients on the waiting list, and to keep patients alive until resolution of the primary dysfunction after graft transplant. There are patients requiring lung transplant in Brazil who do not even come to the point of being referred to a transplant center because there are only seven such centers active in the country. It is urgent to create new centers capable of performing lung transplantation to provide patients with some advanced forms of lung disease a chance to live longer and with better quality of life.
Collapse
|
50
|
The Role of Lysophosphatidic Acid on Airway Epithelial Cell Denudation in a Murine Heterotopic Tracheal Transplant Model. Transplant Direct 2015; 1:e35. [PMID: 27500235 PMCID: PMC4946481 DOI: 10.1097/txd.0000000000000542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/15/2015] [Indexed: 01/06/2023] Open
Abstract
Supplemental digital content is available in the text. Background Chronic rejection is the major leading cause of morbidity and mortality after lung transplantation. Obliterative bronchiolitis (OB), a fibroproliferative disorder of the small airways, is the main manifestation of chronic lung allograft rejection. However, there is currently no treatment for the disease. We hypothesized that lysophosphatidic acid (LPA) participates in the progression of OB. The aim of this study was to reveal the involvement of LPA on the lesion of OB. Methods Ki16198, an antagonist specifically for LPA1 and LPA3, was daily administered into the heterotopic tracheal transplant model mice at the day of transplantation. At days 10 and 28, the allografts were isolated and evaluated histologically. The messenger RNA levels of LPAR in microdissected mouse airway regions were assessed to reveal localization of lysophosphatidic acid receptors. The human airway epithelial cell was used to evaluate the mechanism of LPA-induced suppression of cell adhesion to the extracellular matrix (ECM). Results The administration of Ki16198 attenuated airway epithelial cell loss in the allograft at day 10. Messenger RNAs of LPA1 and LPA3 were detected in the airway epithelial cells of the mice. Lysophosphatidic acid inhibited the attachment of human airway epithelial cells to the ECM and induced cell detachment from the ECM, which was mediated by LPA1 and Rho-kinase pathway. However, Ki16198 did not prevent obliteration of allograft at day 28. Conclusions The LPA signaling is involved in the status of epithelial cells by distinct contribution in 2 different phases of the OB lesion. This finding suggests a role of LPA in the pathogenesis of OB.
Collapse
|