1
|
Jamali Z, Razipour M, Zargar M, Berenji HG, Akrami SM. Ovarian cancer extracellular vesicle biomarkers. Clin Chim Acta 2024; 565:120011. [PMID: 39437983 DOI: 10.1016/j.cca.2024.120011] [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: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Ovarian cancer (OC) remains a significant women's health concern due to its high mortality rate and the challenges posed by late detection. Exploring novel biomarkers could lead to earlier, more specific diagnoses and improved survival rates for OC patients. This review focuses on biomarkers associated with extracellular vesicles (EVs) found in various proximal fluids, including urine, ascites, utero-tubal lavage fluid of OC patients. We highlight these proximal fluids as rich sources of potential biomarkers. The review explains the roles of EV biomarkers in ovarian cancer progression and discusses EV-related proteins and miRNAs as potential diagnostic or prognostic indicators and therapeutic targets. Finally, we highlighted the limitations of examining proximal fluids as sources of biomarkers and encourage researchers to proactively pursue innovative solutions to overcome these challenges.
Collapse
Affiliation(s)
- Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Razipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zargar
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hojat Ghasemnejad Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Seyed Mohammad Akrami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Boster JM, Moore Iii WJ, Stoffel ST, Anderson JT, Gonzales MA, Houle MC, Walter RJ, Morris MJ. Bronchoalveolar Lavage Fluid Cytology of Deployed Military Personnel With Chronic Respiratory Symptoms From the STAMPEDE III Study. Mil Med 2024:usae056. [PMID: 38430524 DOI: 10.1093/milmed/usae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/08/2024] [Indexed: 03/04/2024] Open
Abstract
INTRODUCTION Deployed military personnel may be at risk for developing acute and chronic lung disease. Prior studies of this patient population have revealed that unexplained exertional dyspnea is the most common diagnosis despite an extensive evaluation. There is a concern that an occult disorder may be affecting this population. This study evaluated the role for bronchoalveolar lavage (BAL) fluid analysis in the evaluation of chronic deployment-associated dyspnea. MATERIALS AND METHODS Military personnel who reported chronic respiratory symptoms were evaluated as part of the Study of Active Duty Military for Pulmonary Disease Related to Environmental Deployment Exposures III study. Participants underwent bronchoscopy with BAL as part of a standardized evaluation. RESULTS A total of 308 patients with a mean age of 38 ± 8.6 years underwent bronchoscopy with BAL. BAL cell-count percentages of macrophages, lymphocytes, neutrophils, and eosinophils were: 76.2 ± 17.0%, 16.3 ± 13.4%, 6.6 ± 8.9%, and 0.9 ± 3.2%, respectively. There was no clear differentiation between groups based on increases in lymphocyte counts (P = .640), although lymphocyte values were more elevated (21.4 ± 12.1%) in the interstitial lung disease category. Neutrophil counts (6.6 ± 8.9%) were elevated compared to the reported normal reference values and were increased in the isolated pulmonary function test abnormality (9.4 ± 11.6%), large airway disorder (10.0 ± 7.5%), miscellaneous (10.9 ± 20.2%), and obstructive lung disease (11.0 ± 15.6%) groups. Eosinophil counts were within normal limits (0.9 ± 3.2%) and showed no differences between groups (P = .545); asthma patients trended higher (1.6 ± 5.7%). BAL counts for the exertional dyspnea group were within normal reference values and showed no differences from the entire cohort. CONCLUSIONS The addition of BAL cytology did not help differentiate those patients with unexplained dyspnea from other etiologies.
Collapse
Affiliation(s)
- Joshua M Boster
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - William J Moore Iii
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Steven T Stoffel
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Jess T Anderson
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Michael A Gonzales
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Mateo C Houle
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Robert J Walter
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Michael J Morris
- Pulmonary/Critical Service, Department of Medicine, Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| |
Collapse
|
3
|
de Silva TA, Apte S, Voisey J, Spann K, Tan M, Chambers D, O'Sullivan B. Immunological Landscapes in Lung Transplantation: Insights from T Cell Profiling in BAL and PBMC. Int J Mol Sci 2024; 25:2476. [PMID: 38473722 DOI: 10.3390/ijms25052476] [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: 11/03/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
Lung transplant recipients frequently encounter immune-related complications, including chronic lung allograft dysfunction (CLAD). Monitoring immune cells within the lung microenvironment is pivotal for optimizing post-transplant outcomes. This study examined the proportion of T cell subsets in paired bronchoalveolar lavage (BAL) and peripheral PBMC comparing healthy (n = 4) and lung transplantation patients (n = 6, no CLAD and n = 14 CLAD) using 14-color flow cytometry. CD4+ T cell proportions were reduced in CD3 cells in both PBMC and BAL, and positive correlations were discerned between T cell populations in peripheral PBMC and BAL, suggesting the prospect of employing less invasive PBMC sampling as a means of monitoring lung T cells. Furthermore, regulatory T cells (Tregs) were enriched in BAL when compared to peripheral PBMC for transplant recipients. A parallel positive correlation emerged between Treg proportions in BAL and peripheral PBMC, underscoring potential avenues for monitoring lung Tregs. Finally, the most promising biomarker was the Teff (CD8+Granzyme B+)-Treg ratio, which was higher in both the PBMC and BAL of transplant recipients compared to healthy individuals, and increased in the patients with CLAD compared to no CLAD and healthy patients. Conclusions: Distinct T cell profiles in BAL and peripheral PBMC underscore the significance of localized immune monitoring in lung transplantation. The Teff (CD8+granzyme B+)-Treg ratio, particularly within the context of CLAD, emerges as a promising blood and BAL biomarker reflective of inflammation and transplant-related complications. These findings emphasize the imperative need for personalized immune monitoring strategies that tailored to address the unique immunological milieu in post-transplant lungs.
Collapse
Affiliation(s)
- Tharushi Ayanthika de Silva
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
| | - Simon Apte
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Joanne Voisey
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Maxine Tan
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Daniel Chambers
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| | - Brendan O'Sullivan
- Centre for Genomics and Personalised Health, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
- Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Brisbane, QLD 4001, Australia
- Facility of Clinical Medicine, The University of Queensland, Brisbane, QLD 4001, Australia
| |
Collapse
|
4
|
Tissot A, Durand E, Goronflot T, Coiffard B, Renaud-Picard B, Roux A, Demant X, Mornex JF, Falque L, Salpin M, Le Pavec J, Villeneuve T, Boussaud V, Knoop C, Magnan A, Lair D, Berthelot L, Danger R, Brouard S. Blood MMP-9 measured at 2 years after lung transplantation as a prognostic biomarker of chronic lung allograft dysfunction. Respir Res 2024; 25:88. [PMID: 38336710 PMCID: PMC10858575 DOI: 10.1186/s12931-024-02707-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Long-term outcomes of lung transplantation (LTx) remain hampered by chronic lung allograft dysfunction (CLAD). Matrix metalloproteinase 9 (MMP-9) is a secretory endopeptidase identified as a key mediator in fibrosis processes associated with CLAD. The objective of this study was to investigate whether plasma MMP9 levels may be prognostic of CLAD development. METHODS Participants were selected from the Cohort in Lung Transplantation (COLT) for which a biocollection was associated. We considered two time points, year 1 (Y1) and year 2 (Y2) post-transplantation, for plasma MMP-9 measurements. We analysed stable recipients at those time points, comparing those who would develop a CLAD within the 2 years following the measurement to those who would remain stable 2 years after. RESULTS MMP-9 levels at Y1 were not significantly different between the CLAD and stable groups (230 ng/ml vs. 160 ng/ml, p = 0.4). For the Y2 analysis, 129 recipients were included, of whom 50 developed CLAD within 2 years and 79 remained stable within 2 years. MMP-9 plasma median concentrations were higher in recipients who then developed CLAD than in the stable group (230 ng/ml vs. 118 ng/ml, p = 0.003). In the multivariate analysis, the Y2 MMP-9 level was independently associated with CLAD, with an average increase of 150 ng/ml (95% CI [0-253], p = 0.05) compared to that in the stable group. The Y2 ROC curve revealed a discriminating capacity of blood MMP-9 with an area under the curve of 66%. CONCLUSION Plasmatic MMP-9 levels measured 2 years after lung transplantation have prognostic value for CLAD.
Collapse
Affiliation(s)
- Adrien Tissot
- CHU Nantes, INSERM, Service de Pneumologie, l'institut du thorax, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes Université, 44093, Nantes, France.
| | - Eugénie Durand
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Thomas Goronflot
- CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, Nantes Université, Nantes, France
| | - Benjamin Coiffard
- Department of Respiratory Medicine and Lung Transplantation, APHM, Hôpital Nord, Aix Marseille Univ, Marseille, France
| | - Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, Inserm UMR 1260, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - Xavier Demant
- Service de Pneumologie, Centre Hospitalier Universitaire de Bordeaux, Pessac, France
| | - Jean-François Mornex
- Université Lyon 1, PSL, EPHE, INRAE, IVPC, Hospices Civils de Lyon, groupement hospitalier est, service de pneumologie, Orphalung, RESPIFIL, Université de Lyon, Lyon, France
| | - Loïc Falque
- Service Hospitalier Universitaire de Pneumologie et Physiologie, CHU Grenoble Alpes, Pôle Thorax et Vaisseaux, Grenoble, France
| | - Mathilde Salpin
- APHP Nord-Université Paris Cité, Hôpital Bichat, Service de Pneumologie B et Transplantation Pulmonaire, PHERE UMRS 1152, Université Paris Cité, Paris, France
| | - Jérôme Le Pavec
- Service de Pneumologie et Transplantation Pulmonaire, Groupe hospitalier Marie-Lannelongue -Saint Joseph, Le Plessis-Robinson, Université Paris-Saclay, UMR_S 999, INSERM, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Thomas Villeneuve
- CHU Toulouse, Service de Pneumologie, Université Toulouse III-Paul Sabatier, Toulouse, France
| | | | | | - Antoine Magnan
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - David Lair
- CHU Nantes, Nantes Université, Institut du Thorax, Lung O2, Nantes, France
| | - Laureline Berthelot
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Richard Danger
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Sophie Brouard
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| |
Collapse
|
5
|
Boutin CA, Ison MG. Systematic fungal biomarker or polymerase chain reaction testing in lung transplant recipients: Retrospective analysis to optimize their use. Transpl Infect Dis 2023; 25:e14136. [PMID: 37608632 PMCID: PMC10798658 DOI: 10.1111/tid.14136] [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: 06/12/2023] [Revised: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Among lung transplant recipients, serial bronchoscopies are performed frequently. Often, serial galactomannan (GM), 1,3-β-d-glucan (BDG), and Pneumocystis jirovecii (PJ) testing is performed with these broncho-alveolar lavages (BALs) as standard of care with limited data to support their routine use. METHODS After Institutional Review Board approval, we retrospectively collected all blood and BAL GM, BDG, and PJ test results from January 2015 to July 20, 2022. Primary data collection from the Northwestern Medicine EDW was supplemented by manual chart review. RESULTS During the study period, 236 lung transplant recipients were cared for by our center. Of these patients, 217 (91.9%) had 1418 GM tests performed; 61 (4.3%) were positive (index ≥1). Fungal cultures were requested for most BAL-GM (90.7%). Out of duplicates in same BAL, results discrepancy was minimal (3.4%). 172 (72.9%) had BDG tests were performed; 25.6% were positive. Thirteen patients had multiple BDG during one hospitalization (mean 2.3 tests); none of the negative test repeated became positive. Eleven negative BDG were seen in patients with invasive aspergillosis (IA). Note that, 577 PJ testing were performed (direct fluorescent antibody [n = 494] or polymerase chain reaction [PCR] [n = 80], or both [n = 3]) in 174 different patients. None were positive. CONCLUSION Despite supplemental GM, BDG, and Pneumocystis jirovecii pneumonia PCR being performed routinely on lung transplant recipients undergoing BAL at our center, the data suggests a more tailored approach may be appropriate. There is no role for routine serial testing with these assays during a single hospitalization. BDG confers no added-value over GM with cultures for IA diagnosis.
Collapse
Affiliation(s)
- Catherine-Audrey Boutin
- Department of Medicine, Divisions of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medicine, Division of Infectious Disease, University of Montreal Hospital Center, Montreal, Canada
| | - Michael G Ison
- Respiratory Diseases Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, Rockville, Maryland, USA
| |
Collapse
|
6
|
Ravichandran R, Itabashi Y, Zhou F, Lin Y, Mohanakumar T, Chapman WC. Circulating exosomes from brain death and cardiac death donors have distinct molecular and immunologic properties: A pilot study. Clin Transplant 2023; 37:e15067. [PMID: 37428019 PMCID: PMC11019898 DOI: 10.1111/ctr.15067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND AIMS Comparison of donation after brain death (DBD) and donation after cardiac death (DCD) lung tissue before transplantation have demonstrated activation of pro-inflammatory cytokine pathway in DBD donors. The molecular and immunological properties of circulating exosomes from DBD and DCD donors were not previously described. METHODS We collected plasma from 18 deceased donors (12 DBD and six DCD). Cytokines were analyzed by 30-Plex luminex Panels. Exosomes were analyzed for liver self-antigen (SAg), Transcription Factors and HLA class II (HLA-DR/DQ) using western blot. C57BL/6 animals were immunized with isolated exosomes to determine strength and magnitude of immune responses. Interferon (IFN)-γ and tumor necrosis factor-α producing cells were quantified by ELISPOT, specific antibodies to HLA class II antigens were measured by ELISA RESULTS: We demonstrate increased plasma levels of IFNγ, EGF, EOTAXIN, IP-10, MCP-1, RANTES, MIP-β, VEGF, and interleukins - 6/8 in DBD plasma versus DCD. MiRNA isolated from exosome of DBD donors demonstrated significant increase in miR-421, which has been reported to correlate with higher level of Interleukin-6. Higher levels of liver SAg Collagen III (p = .008), pro-inflammatory transcription factors (NF-κB, p < .05; HIF1α, p = .021), CIITA (p = .011), and HLA class II (HLA-DR, p = .0003 and HLA-DQ, p = .013) were detected in exosomes from DBD versus DCD plasma. The circulating exosomes isolated from DBD donors were immunogenic in mice and led to the development of Abs to HLA-DR/DQ. CONCLUSIONS This study provides potential new mechanisms by which DBD organs release exosomes that can activate immune pathways leading to cytokine release and allo-immune response.
Collapse
Affiliation(s)
| | - Yoshihiro Itabashi
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Fangyu Zhou
- Division of General Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | - Yiing Lin
- Division of General Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | | | - William C. Chapman
- Division of General Surgery, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
7
|
Armati M, Cattelan S, Guerrieri M, Messina M, Perea B, Genovese M, d'Alessandro M, Gangi S, Cameli P, Perillo F, Bennett D, Fossi A, Bargagli E, Bergantini L. Collagen Type IV Alpha 5 Chain in Bronchiolitis Obliterans Syndrome After Lung Transplant: The First Evidence. Lung 2023; 201:363-369. [PMID: 37402896 PMCID: PMC10444639 DOI: 10.1007/s00408-023-00632-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023]
Abstract
INTRODUCTION Bronchiolitis obliterans syndrome (BOS) is the most common form of CLAD and is characterized by airflow limitation and an obstructive spirometry pattern without parenchymal opacities. The protein signature of BOS lesions concerns extracellular matrix organization and aberrant basement membrane composition. In this pilot study, we investigated the presence of COL4A5 in the serum of patients with BOS. METHODS 41 patients who had undergone LTX were enrolled. Of these, 27 developed BOS and 14 (control group) were considered stable at the time of serum sampling. Of BOS patients, serum samples were analysed at the time of BOS diagnosis and before the clinical diagnosis (pre-BOS). COL4A5 levels were detected through the ELISA kit. RESULTS Serum concentrations of COL4A5 were higher in pre-BOS than in stable patients (40.5 ± 13.9 and 24.8 ± 11.4, respectively, p = 0.048). This protein is not influenced by comorbidities, such as acute rejection or infections, or by therapies. Survival analysis also reveals that a higher level of COL4A5 was also associated with less probability of survival. Our data showed a correlation between concentrations of COL4A5 and FEV1 at the time of diagnosis of BOS. CONCLUSION Serum concentrations of COL4A5 can be considered a good prognostic marker due to their association with survival and correlation with functional parameters.
Collapse
Affiliation(s)
- M Armati
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - S Cattelan
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - M Guerrieri
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - M Messina
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - B Perea
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - M Genovese
- Unit of Respiratory Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - M d'Alessandro
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - S Gangi
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | | | - F Perillo
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - D Bennett
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - A Fossi
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - E Bargagli
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy
| | - L Bergantini
- Department of Medical Sciences, Surgery and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, 53100, Siena, Italy.
| |
Collapse
|
8
|
Markers of Bronchiolitis Obliterans Syndrome after Lung Transplant: Between Old Knowledge and Future Perspective. Biomedicines 2022; 10:biomedicines10123277. [PMID: 36552035 PMCID: PMC9775233 DOI: 10.3390/biomedicines10123277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the most common form of CLAD and is characterized by airflow limitation and an obstructive spirometric pattern without high-resolution computed tomography (HRCT) evidence of parenchymal opacities. Computed tomography and microCT analysis show abundant small airway obstruction, starting from the fifth generation of airway branching and affecting up to 40-70% of airways. The pathogenesis of BOS remains unclear. It is a multifactorial syndrome that leads to pathological tissue changes and clinical manifestations. Because BOS is associated with the worst long-term survival in LTx patients, many studies are focused on the early identification of BOS. Markers may be useful for diagnosis and for understanding the molecular and immunological mechanisms involved in the onset of BOS. Diagnostic and predictive markers of BOS have also been investigated in various biological materials, such as blood, BAL, lung tissue and extracellular vesicles. The aim of this review was to evaluate the scientific literature on markers of BOS after lung transplant. We performed a systematic review to find all available data on potential prognostic and diagnostic markers of BOS.
Collapse
|
9
|
Moshkelgosha S, Duong A, Wilson G, Andrews T, Berra G, Renaud-Picard B, Liu M, Keshavjee S, MacParland S, Yeung J, Martinu T, Juvet S. Interferon-stimulated and metallothionein-expressing macrophages are associated with acute and chronic allograft dysfunction after lung transplantation. J Heart Lung Transplant 2022; 41:1556-1569. [DOI: 10.1016/j.healun.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
|
10
|
Clinical Validation of a Plasma Donor-derived Cell-free DNA Assay to Detect Allograft Rejection and Injury in Lung Transplant. Transplant Direct 2022; 8:e1317. [PMID: 35372675 PMCID: PMC8963832 DOI: 10.1097/txd.0000000000001317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/04/2022] [Accepted: 02/26/2022] [Indexed: 01/29/2023] Open
|
11
|
Greenland NY, Deiter F, Calabrese DR, Hays SR, Kukreja J, Leard LE, Kolaitis NA, Golden JA, Singer JP, Greenland JR. Inflammation on bronchoalveolar lavage cytology is associated with decreased chronic lung allograft dysfunction-free survival. Clin Transplant 2022; 36:e14639. [PMID: 35246990 DOI: 10.1111/ctr.14639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lung transplant recipients undergo bronchoalveolar lavage (BAL) to detect antecedents of chronic lung allograft dysfunction (CLAD), but routine assessment of BAL cytology is controversial. We hypothesized that inflammation on BAL cytology would predict CLAD-free survival. METHODS In a single-center retrospective cohort, associations between cytology results and clinical characteristics were compared using generalized-estimating equation-adjusted regression. The association between BAL inflammation and CLAD or death risk was assessed using time-dependent Cox models. RESULTS In 3,365 cytology reports from 451 subjects, inflammation was the most common finding (6.2%, 210 cases), followed by fungal forms (5.3%, 178 cases, including 24 cases of suspected Aspergillus). Inflammation on BAL cytology was more common in procedures for symptoms (8.5%) versus surveillance (3.2%, P<0.001). Inflammation on cytology was associated with automated neutrophil and lymphocyte counts, acute cellular rejection, infection, and portended a 2.2-fold hazard ratio (CI 1.2-4.0, P = 0.007) for CLAD or death. However, inflammation by cytology did not inform CLAD-free survival risk beyond automated BAL cell counts (P = 0.57). CONCLUSIONS Inflammation on BAL cytology is clinically significant, suggesting acute rejection or infection and increased risk of CLAD or death. However, other indicators of allograft inflammation can substitute for much of the information provided by BAL cytology. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nancy Y Greenland
- Department of Anatomic Pathology, University of California, San Francisco, USA.,Veterans Affairs Health Care System, San Francisco, California, USA
| | - Fred Deiter
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Daniel R Calabrese
- Veterans Affairs Health Care System, San Francisco, California, USA.,Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Steven R Hays
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, USA
| | - Lorriana E Leard
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Nicholas A Kolaitis
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jeffrey A Golden
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jonathan P Singer
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - John R Greenland
- Veterans Affairs Health Care System, San Francisco, California, USA.,Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| |
Collapse
|
12
|
Ravichandran R, Itabashi Y, Liu W, Bansal S, Rahman M, Poulson C, Fleming T, Bremner RM, Smith M, Mohanakumar T. A decline in club cell secretory proteins in lung transplantation is associated with release of natural killer cells exosomes leading to chronic rejection. J Heart Lung Transplant 2021; 40:1517-1528. [PMID: 34627707 PMCID: PMC11019779 DOI: 10.1016/j.healun.2021.08.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/06/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND In human lung transplant recipients, a decline in club cell secretory protein (CCSP) in bronchoalveolar lavage fluid has been associated with chronic lung allograft dysfunction (CLAD) as well as the induction of exosomes and immune responses that lead to CLAD. However, the mechanisms by which CCSP decline contributes to CLAD remain unknown. METHODS To define the mechanisms leading to CCSP decline and chronic rejection, we employed two mouse models: 1) chronic rejection after orthotopic single lung transplantation and 2) anti-major histocompatibility complex (MHC) class I-induced obliterative airway disease. RESULTS In the chronic rejection mouse model, we detected circulating exosomes with donor MHC (H2b) and lung self-antigens and also development of antibodies to H2b and lung self-antigens and then a decline in CCSP. Furthermore, DBA2 mice that received injections of these exosomes developed antibodies to donor MHC and lung self-antigens. In the chronic rejection mouse model, natural killer (NK) and CD8 T cells were the predominant graft-infiltrating cells on day 14 of rejection followed by exosomes containing NK cell-associated and cytotoxic molecules on day 14 and 28. When NK cells were depleted, exosomes with NK cell-associated and cytotoxic molecules as well as fibrosis decreased. CONCLUSIONS Induction of exosomes led to immune responses to donor MHC and lung self-antigens, resulting in CCSP decline, leading to NK cell infiltration and release of exosomes from NK cells. These results suggest a novel role for exosomes derived from NK cells in the pathogenesis of chronic lung allograft rejection.
Collapse
Affiliation(s)
| | | | - Wei Liu
- Norton Thoracic Institute, Phoenix, Arizona
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Itabashi Y, Ravichandran R, Bansal S, Bharat A, Hachem R, Bremner R, Smith M, Mohanakumar T. Decline in Club Cell Secretory Proteins, Exosomes Induction and Immune Responses to Lung Self-antigens, Kα1 Tubulin and Collagen V, Leading to Chronic Rejection After Human Lung Transplantation. Transplantation 2021; 105:1337-1346. [PMID: 32890135 PMCID: PMC7917153 DOI: 10.1097/tp.0000000000003428] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD), is a major hurdle for long-term lung allograft survival after lung transplant and roughly 50% of lung transplant recipients (LTxRs) develop CLAD within 5 years. The mechanisms of CLAD development remain unknown. Donor-specific immune responses to HLA and lung self-antigens (SAgs) are vital to the pathogenesis of CLAD. Reduction in Club cell secretory protein (CCSP) has been reported in bronchoalveolar lavage (BAL) fluid samples from LTxRs with bronchiolitis obliterans syndrome (BOS). CCSP levels in BAL fluid and development of antibodies to lung SAgs in plasma were determined by ELISA. Cytokines in BAL fluid were analyzed by 30-plex Luminex panel. Exosomes from BAL fluid or plasma were analyzed for SAgs, natural killer (NK) cells markers, and cytotoxic molecules. RESULTS We demonstrate that LTxRs with BOS have lower CCSP levels up to 9 months before BOS diagnosis. LTxRs with antibodies to SAgs 1-year posttransplant also developed DSA (43%) and had lower CCSP. BOS with lower CCSP also induced Interleukin-8 and reduced vascular endothelial growth factor. Exosomes from BOS contained increased SAgs, NK cells markers, and cytotoxic molecules. CONCLUSIONS We conclude lower CCSP leads to inflammation, pro-inflammatory cytokine production, immune responses to HLA and SAgs, and induction of exosomes. For the first time, we demonstrate that CCSP loss results in exosome release from NK cells capable of stimulating innate and adaptive immunity posttransplant. This increases the risk of BOS, suggesting a role of NK cell exosomes in CLAD development.
Collapse
Affiliation(s)
- Yoshihiro Itabashi
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | | | - Sandhya Bansal
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Ankit Bharat
- Department of Surgery, University of Chicago, Chicago, IL, United States
| | - Ramsey Hachem
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ross Bremner
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Michael Smith
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - T. Mohanakumar
- Norton Thoracic Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| |
Collapse
|
14
|
Berra G, Farkona S, Mohammed-Ali Z, Kotlyar M, Levy L, Clotet-Freixas S, Ly P, Renaud-Picard B, Zehong G, Daigneault T, Duong A, Batruch I, Jurisica I, Konvalinka A, Martinu T. Association between renin-angiotensin system and chronic lung allograft dysfunction. Eur Respir J 2021; 58:13993003.02975-2020. [PMID: 33863738 DOI: 10.1183/13993003.02975-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/06/2021] [Indexed: 11/05/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major cause of death after lung transplantation. Angiotensin II (AngII), the main effector of the renin-angiotensin (RA) system, elicits fibrosis in both kidney and lung. We identified 6 AngII-regulated proteins (RHOB, BST1, LYPA1, GLNA, TSP1, LAMB1) increased in urine of patients with kidney allograft fibrosis. We hypothesized that RA system is active in CLAD and that AngII-regulated proteins are increased in bronchoalveolar lavage fluid (BAL) of CLAD patients.We performed immunostaining of AngII receptors (AGTR1 and AGTR2) and TSP1/GLNA in 10 CLAD lungs and 5 controls. Using mass spectrometry, we quantified peptides corresponding to AngII-regulated proteins in BAL of 40 lung transplant recipients (CLAD, stable and acute lung allograft dysfunction (ALAD)). Machine learning algorithms were developed to predict CLAD based on BAL peptide concentrations.Immunostaining demonstrated significantly more AGTR1+ cells in CLAD versus control lungs (p=0.02). TSP1 and GLNA immunostaining positively correlated with the degree of lung fibrosis (R2=0.42 and 0.57, respectively). In BAL, we noted a trend toward higher concentrations of AngII-regulated peptides in patients with CLAD at the time of bronchoscopy, and significantly higher concentrations of BST1, GLNA and RHOB peptides in patients that developed CLAD at follow-up (p<0.05). Support vector machine classifier discriminated CLAD from stable and ALAD patients at the time of bronchoscopy with AUC 0.86, and accurately predicted subsequent CLAD development (AUC 0.97).Proteins involved in the RA system are increased in CLAD lung and BAL. AngII-regulated peptides measured in BAL may accurately identify patients with CLAD and predict subsequent CLAD development.
Collapse
Affiliation(s)
- Gregory Berra
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,First two authors contributed equally
| | - Sofia Farkona
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,First two authors contributed equally
| | - Zahraa Mohammed-Ali
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Toronto, ON, Canada, Canada
| | - Liran Levy
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Phillip Ly
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Benjamin Renaud-Picard
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Guan Zehong
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Tina Daigneault
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Allen Duong
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Ihor Batruch
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum, Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Toronto, ON, Canada, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada .,Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada.,Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Last two authors contributed equally
| | - Tereza Martinu
- Toronto Lung Transplant Program, University Health Network, Toronto, ON, Canada .,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,Last two authors contributed equally
| |
Collapse
|
15
|
Potential novel biomarkers for chronic lung allograft dysfunction and azithromycin responsive allograft dysfunction. Sci Rep 2021; 11:6799. [PMID: 33762606 PMCID: PMC7990920 DOI: 10.1038/s41598-021-85949-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
Chronic Lung Allograft Dysfunction (CLAD), manifesting as Bronchiolitis Obliterans Syndrome (BOS) or Restrictive Allograft Syndrome (RAS), is the main reason for adverse long-term outcome after Lung Transplantation (LTX). Until now, no specific biomarkers exist to differentiate between CLAD phenotypes. Therefore, we sought to find suitable cytokines to distinguish between BOS, RAS and Azithromycin Responsive Allograft Dysfunction (ARAD); and reveal potential similarities or differences to end-stage fibrotic diseases. We observed significantly increased Lipocalin-2 serum concentrations in RAS compared to BOS patients. In addition, in RAS patients immunohistochemistry revealed Lipocalin-2 expression in bronchial epithelium and alveolar walls. Patients with ARAD showed significantly lower Activin-A serum concentrations compared to Stable-LTX and BOS patients. Further, increased serum concentrations of Lipocalin-2 and Activin-A were predictors of worse freedom-from-CLAD in Stable-LTX patients. These biomarkers serve as promising serum biomarkers for CLAD prediction and seem suitable for implementation in clinical practice.
Collapse
|
16
|
Sikkeland LIB, Durheim MT, Riste TB, Kongerud J, Alexis NE, Holm AM. Relation of sputum neutrophilia to the development of chronic lung allograft dysfunction after lung transplantation. Clin Transplant 2021; 35:e14188. [PMID: 33315265 DOI: 10.1111/ctr.14188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 11/29/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is a serious complication after lung transplantation (LuTx) and is associated with elevated proportions of neutrophils in bronchoalveolar lavage (BAL). Induced sputum is a less-invasive sampling method than BAL and assesses markers of inflammation on the surfaces of large central airways. We wanted to examine whether % neutrophil levels in induced sputum were elevated prior to CLAD diagnosis among LuTx recipients, and whether sputum markers of inflammation can be used as a tool for predicting the development of CLAD. Induced sputum samples were collected at 1, 3, 6, 12, and 24 months post-LuTx in 36 patients with a history of COPD or pulmonary fibrosis, and of these, 16 developed CLAD either during or after the sputum surveillance period. At 2 years, median (IQR) % neutrophils in induced sputum were significantly higher among patients with CLAD compared with those without CLAD [73 (52-80) % vs 59 (41-76) %, p = .01]. Interestingly, we found a significant increase in the rate of change in % neutrophils beginning at 90 days preceding the diagnosis of CLAD. This suggests using sputum neutrophil percentage as a surveillance modality for monitoring lung allograft function after LuTx.
Collapse
Affiliation(s)
- Liv Ingunn Bjoner Sikkeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Respiratory Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Michael Thomas Durheim
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Respiratory Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tonje Bøyum Riste
- Department of Respiratory Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Johny Kongerud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Respiratory Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Neil E Alexis
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Are Martin Holm
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Respiratory Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
17
|
Establishment of a New PNA-FISH Method for Aspergillus fumigatus Identification: First Insights for Future Use in Pulmonary Samples. Microorganisms 2020; 8:microorganisms8121950. [PMID: 33316925 PMCID: PMC7763223 DOI: 10.3390/microorganisms8121950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/28/2020] [Accepted: 12/05/2020] [Indexed: 01/01/2023] Open
Abstract
Aspergillus fumigatus is the main causative agent of Invasive Aspergillosis. This mold produces conidia that when inhaled by immunocompromized hosts can be deposited in the lungs and germinate, triggering disease. In this paper, the development of a method using peptide nucleic acid-fluorescence in situ hybridization (PNA-FISH) is described. The PNA-FISH probe was tested in several strains and a specificity and sensitivity of 100% was obtained. Detection of A. fumigatussensu stricto was then achieved in artificial sputum medium (ASM) pre-inoculated with 1 × 102 conidia·mL-1-1 × 103 conidia·mL-1, after a germination step of 24 h. The PNA-FISH method was evaluated in 24 clinical samples (10 sputum, 8 bronchoalveolar lavage (BAL), and 6 bronchial lavage (BL)) that were inoculated with 1 × 104 conidia·mL-1 in sputum; 1 × 103 conidia·mL-1 in BL and BAL, for 24 h. Despite a specificity of 100%, the sensitivity was 79%. This relatively low sensitivity can be explained by the fact that hyphae can yield "fungal ball" clusters, hindering pipetting procedures and subsequent detection, leading to false negative results. Nonetheless, this study showed the potential of the PNA-FISH method for A. fumigatussensu stricto detection since it takes only 1 h 30 m to perform the procedure with a pre-enrichment step of 6 h (pure cultures) and 24 h (clinical samples), and might provide a suitable alternative to the existing methods for studies in pure cultures and in clinical settings.
Collapse
|
18
|
Martinu T, Koutsokera A, Benden C, Cantu E, Chambers D, Cypel M, Edelman J, Emtiazjoo A, Fisher AJ, Greenland JR, Hayes D, Hwang D, Keller BC, Lease ED, Perch M, Sato M, Todd JL, Verleden S, von der Thüsen J, Weigt SS, Keshavjee S. International Society for Heart and Lung Transplantation consensus statement for the standardization of bronchoalveolar lavage in lung transplantation. J Heart Lung Transplant 2020; 39:1171-1190. [PMID: 32773322 PMCID: PMC7361106 DOI: 10.1016/j.healun.2020.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 01/09/2023] Open
Abstract
Bronchoalveolar lavage (BAL) is a key clinical and research tool in lung transplantation (LTx). However, BAL collection and processing are not standardized across LTx centers. This International Society for Heart and Lung Transplantation-supported consensus document on BAL standardization aims to clarify definitions and propose common approaches to improve clinical and research practice standards. The following 9 areas are covered: (1) bronchoscopy procedure and BAL collection, (2) sample handling, (3) sample processing for microbiology, (4) cytology, (5) research, (6) microbiome, (7) sample inventory/tracking, (8) donor bronchoscopy, and (9) pediatric considerations. This consensus document aims to harmonize clinical and research practices for BAL collection and processing in LTx. The overarching goal is to enhance standardization and multicenter collaboration within the international LTx community and enable improvement and development of new BAL-based diagnostics.
Collapse
Affiliation(s)
- Tereza Martinu
- Toronto Lung Transplant Program, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| | - Angela Koutsokera
- Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Lung Transplant Program, Division of Pulmonology, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Edward Cantu
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Chambers
- Lung Transplant Program, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Marcelo Cypel
- Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey Edelman
- Lung Transplant Program, Puget Sound VA Medical Center, Seattle, Washington
| | - Amir Emtiazjoo
- Lung Transplant Program, University of Florida, Gainesville, Florida
| | - Andrew J Fisher
- Institute of Transplantation, Newcastle Upon Tyne Hospitals and Newcastle University, United Kingdom
| | - John R Greenland
- Department of Medicine, VA Health Care System, San Francisco, California
| | - Don Hayes
- Lung Transplant Program, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - David Hwang
- Department of Pathology, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Brian C Keller
- Lung Transplant Program, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Erika D Lease
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Michael Perch
- Lung Transplant Program, Rigshospitalet, Copenhagen, Denmark
| | - Masaaki Sato
- Department of Surgery, University of Tokyo, Tokyo, Japan
| | - Jamie L Todd
- Lung Transplant Program, Duke University Medical Center, Durham, North Carolina
| | - Stijn Verleden
- Laboratory of Pneumology, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - S Samuel Weigt
- Lung Transplant Program, University of California Los Angeles, Los Angeles, California
| | - Shaf Keshavjee
- Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
19
|
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
|
20
|
Yang S, Abuduwufuer A, Lv W, Bao F, Hu J. [Predictors for the Bronchiolitis Obliterans Syndrome in Lung Transplant Patient]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:496-502. [PMID: 32517455 PMCID: PMC7309540 DOI: 10.3779/j.issn.1009-3419.2020.101.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
肺移植是治疗终末期肺病的有效方法。目前,肺移植术后1年生存率已达到80%,由于闭塞性细支气管炎综合症(bronchiolitis obliterans syndrome, BOS)的发生,5年生存率维持在50%左右。BOS是一个纤维化的过程,最终导致不可逆的气道闭塞。缺血-再灌注损伤、感染、氧化应激以及急性排斥反应等多个因素参与了BOS的发生。研究证实BOS的早期诊断与预后良好相关。因此,寻找灵敏、特异的BOS预测标记物对于提高肺移植患者长期生存具有重要的科学和临床意义。本文就与BOS发生发展相关的免疫调节细胞、分泌性蛋白质、细胞膜蛋白等指标的变化在BOS早期诊断中的作用进行综述。
Collapse
Affiliation(s)
- Sijia Yang
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | - Wang Lv
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Feichao Bao
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Hu
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
21
|
Zhang CYK, Ahmed M, Huszti E, Levy L, Hunter SE, Boonstra KM, Moshkelgosha S, Sage AT, Azad S, Zamel R, Ghany R, Yeung JC, Crespin OM, Frankel C, Budev M, Shah P, Reynolds JM, Snyder LD, Belperio JA, Singer LG, Weigt SS, Todd JL, Palmer SM, Keshavjee S, Martinu T. Bronchoalveolar bile acid and inflammatory markers to identify high-risk lung transplant recipients with reflux and microaspiration. J Heart Lung Transplant 2020; 39:934-944. [PMID: 32487471 DOI: 10.1016/j.healun.2020.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/20/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Gastroesophageal reflux disease (GERD) is a risk factor for chronic lung allograft dysfunction. Bile acids-putative markers of gastric microaspiration-and inflammatory proteins in the bronchoalveolar lavage (BAL) have been associated with chronic lung allograft dysfunction, but their relationship with GERD remains unclear. Although GERD is thought to drive chronic microaspiration, the selection of patients for anti-reflux surgery lacks precision. This multicenter study aimed to test the association of BAL bile acids with GERD, lung inflammation, allograft function, and anti-reflux surgery. METHODS We analyzed BAL obtained during the first post-transplant year from a retrospective cohort of patients with and without GERD, as well as BAL obtained before and after Nissen fundoplication anti-reflux surgery from a separate cohort. Levels of taurocholic acid (TCA), glycocholic acid, and cholic acid were measured using mass spectrometry. Protein markers of inflammation and injury were measured using multiplex assay and enzyme-linked immunosorbent assay. RESULTS At 3 months after transplantation, TCA, IL-1β, IL-12p70, and CCL5 were higher in the BAL of patients with GERD than in that of no-GERD controls. Elevated TCA and glycocholic acid were associated with concurrent acute lung allograft dysfunction and inflammatory proteins. The BAL obtained after anti-reflux surgery contained reduced TCA and inflammatory proteins compared with that obtained before anti-reflux surgery. CONCLUSIONS Targeted monitoring of TCA and selected inflammatory proteins may be useful in lung transplant recipients with suspected reflux and microaspiration to support diagnosis and guide therapy. Patients with elevated biomarker levels may benefit most from anti-reflux surgery to reduce microaspiration and allograft inflammation.
Collapse
Affiliation(s)
- Chen Yang Kevin Zhang
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Musawir Ahmed
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Liran Levy
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Sarah E Hunter
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Kristen M Boonstra
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Sajad Moshkelgosha
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Andrew T Sage
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Sassan Azad
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Ricardo Zamel
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Rasheed Ghany
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Jonathan C Yeung
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Oscar M Crespin
- Division of General Surgery, University Health Network, University of Toronto, Toronto, Canada
| | | | | | - Pali Shah
- Johns Hopkins University Hospital, Baltimore, Maryland
| | | | | | | | - Lianne G Singer
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | | | - Jamie L Todd
- Duke University Medical Center, Durham, North Carolina
| | | | - Shaf Keshavjee
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Tereza Martinu
- Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada.
| | | |
Collapse
|
22
|
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
|
23
|
Kelly FL, Weinberg KE, Nagler AE, Nixon AB, Star MD, Todd JL, Brass DM, Palmer SM. EGFR-Dependent IL8 Production by Airway Epithelial Cells After Exposure to the Food Flavoring Chemical 2,3-Butanedione. Toxicol Sci 2020; 169:534-542. [PMID: 30851105 DOI: 10.1093/toxsci/kfz066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
2,3-Butanedione (DA), a component of artificial butter flavoring, is associated with the development of occupational bronchiolitis obliterans (BO), a disease of progressive airway fibrosis resulting in lung function decline. Neutrophilic airway inflammation is a consistent feature of BO across a range of clinical contexts and may contribute to disease pathogenesis. Therefore, we sought to determine the importance of the neutrophil chemotactic cytokine interleukin-8 (IL-8) in DA-induced lung disease using in vivo and in vitro model systems. First, we demonstrated that levels of Cinc-1, the rat homolog of IL-8, are increased in the lung fluid and tissue compartment in a rat model of DA-induced BO. Next, we demonstrated that DA increased IL-8 production by the pulmonary epithelial cell line NCI-H292 and by primary human airway epithelial cells grown under physiologically relevant conditions at an air-liquid interface. We then tested the hypothesis that DA-induced epithelial IL-8 protein occurs in an epidermal growth factor receptor (EGFR)-dependent manner. In these in vitro experiments we demonstrated that epithelial IL-8 protein is blocked by the EGFR tyrosine kinase inhibitor AG1478 and by inhibition of tumor necrosis factor-alpha converting enzyme using the small molecule inhibitor, TAPI-1. Finally, we demonstrated that DA-induced IL-8 is dependent upon ERK1/2 and Mitogen activated protein kinase kinase activation downstream of EGFR signaling using the small molecule inhibitors AG1478 and PD98059. Together these novel in vivo and in vitro observations support that EGFR-dependent IL-8 production occurs in DA-induced BO. Further studies are warranted to determine the importance of IL-8 in BO pathogenesis.
Collapse
Affiliation(s)
- Francine L Kelly
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| | - Kaitlyn E Weinberg
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| | - Andrew E Nagler
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| | - Andrew B Nixon
- Division of Oncology, Duke University Medical Center, Durham, NC 27710
| | - Mark D Star
- Division of Oncology, Duke University Medical Center, Durham, NC 27710
| | - Jamie L Todd
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| | - David M Brass
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| | - Scott M Palmer
- Division of Pulmonary and Critical Care medicine, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
24
|
Abstract
Introduction: Lung transplantation remains an important treatment for patients with end stage lung disease. Chronic lung allograft dysfunction (CLAD) remains the greatest limiting factor for long term survival. As the diagnosis of CLAD is based on pulmonary function tests, significant lung injury is required before a diagnosis is feasible, likely when irreversible damage has already occurred. Therefore, research is ongoing for early CLAD recognition, with biomarkers making up a substantial amount of this research.Areas covered: The purpose of this review is to describe available biomarkers, focusing on those which aid in predicting CLAD and distinguishing between different CLAD phenotypes. We describe biomarkers presenting in bronchial alveolar lavage (BAL) as well as circulating in peripheral blood, both of which offer an appealing alternative to lung biopsy.Expert opinion: Development of CLAD involves complex, multiple immune and nonimmune mechanisms. Therefore, evaluation of potential CLAD biomarkers serves a dual purpose: clinically, the goal remains early detection and identification of patients at increased risk. Simultaneously, biomarkers offer insight into the different mechanisms involved in the pathophysiology of CLAD, leading to the development of possible interventions. The ultimate goal is the development of both preventive and early intervention strategies for CLAD to improve the overall survival of our lung transplant recipients.
Collapse
Affiliation(s)
- Osnat Shtraichman
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Pulmonary institute, Rabin Medical Center, Petach Tikva, Israel; Sackler School of Medicine, Tel Aviv, Israel
| | - Joshua M Diamond
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Todd JL, Kelly FL, Nagler A, Banner K, Pavlisko EN, Belperio JA, Brass D, Weigt SS, Palmer SM. Amphiregulin contributes to airway remodeling in chronic allograft dysfunction after lung transplantation. Am J Transplant 2020; 20:825-833. [PMID: 31665560 PMCID: PMC7042065 DOI: 10.1111/ajt.15667] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/04/2019] [Accepted: 10/17/2019] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD), a condition of excess matrix deposition and airways fibrosis, limits survival after lung transplantation. Amphiregulin (Areg) is an epidermal growth factor receptor (EGFR) ligand suggested to regulate airway injury and repair. We sought to determine whether Areg expression increases in CLAD, localize the cellular source of Areg induction in CLAD, and assess its effects on airway matrix deposition. Lung fluid Areg protein was quantified in patients with or without CLAD. In situ hybridization was performed to localize Areg and EGFR transcript in CLAD and normal lung tissue. Expression of hyaluronan, a matrix constituent that accumulates in CLAD, was measured in Areg-exposed bronchial epithelial cells in the presence or absence of an EGFR inhibitor. We demonstrated that lung fluid Areg protein was significantly increased in CLAD in a discovery and replication cohort. Areg and EGFR transcripts were abundantly expressed within CLAD tissue, localized to basally distributed airway epithelial cells overlying fibrotic regions. Areg-exposed bronchial epithelial cells increased hyaluronan and hyaluronan synthase expression in an EGFR-dependent manner. Collectively, these novel observations suggest that Areg contributes to airway remodeling and CLAD. Moreover these data implicate a role for EGFR signaling in CLAD pathogenesis, suggesting novel therapeutic targets.
Collapse
Affiliation(s)
- Jamie L. Todd
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina,Duke Clinical Research Institute; Duke University Medical Center; Durham, North Carolina
| | - Fran L. Kelly
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina
| | - Andrew Nagler
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina
| | - Kane Banner
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina
| | | | - John A. Belperio
- University of California Los Angeles; Department of Medicine; Division of Pulmonary Medicine; Los Angeles, California
| | - David Brass
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina
| | - S. Sam Weigt
- University of California Los Angeles; Department of Medicine; Division of Pulmonary Medicine; Los Angeles, California
| | - Scott M. Palmer
- Duke University Medical Center; Department of Medicine; Division of Pulmonary, Allergy and Critical Care Medicine; Durham, North Carolina,Duke Clinical Research Institute; Duke University Medical Center; Durham, North Carolina
| |
Collapse
|
26
|
Li Y, Shu P, Tang L, Yang X, Fan J, Zhang X. FK506 combined with GM6001 prevents tracheal obliteration in a mouse model of heterotopic tracheal transplantation. Transpl Immunol 2019; 57:101244. [PMID: 31526865 DOI: 10.1016/j.trim.2019.101244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/08/2019] [Accepted: 09/13/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Obliterative bronchiolitis (OB) is the major complication limiting the long-term survival of allografts after lung transplantation. In this study, we investigated the effect of tacrolimus (FK506) combined with GM6001,a matrix metalloproteinase (MMP) inhibitor, on the formation of OB using a mouse heterotopic tracheal transplantation model. METHODS Syngeneic tracheal grafts were transplanted heterotopically from BALB/c mice to BALB/c mice. Allografts from C57BL/6 mice were transplanted to BALB/c mice. Isograft group, allograft group, allograft+FK506 group, allograft +GM6001 group and allograft+FK506 + GM6001 group was given respectively intraperitoneal injection of saline, saline, FK506, GM6001 and FK506 + GM6001 once a day. At 28 day after transplantation, OB incidence was determined by hematoxylin-eosin staining and the expressions of MMPs and cytokines were assessed using enzyme linked immunosorbent assay, immunohistochemical assays and western blot assay. RESULTS The tracheal occlusion rates of isograft group, allograft group, allograft+FK506 group, allograft+GM6001 group and allograft+FK506 + GM6001 group were 0, 74.1 ± 9.79%, 34.4 ± 6.04%, 40.3 ± 8.77% and 26.5 ± 5.73% respectively. There were significant differences between the latter two groups (P < .001). The serum MMP-8 and MMP-9 levels of allograft group were significantly higher than those of isograft group (P < .05) and had no significant decrease when treated by FK506. The serum MMP-8 and MMP-9 levels of allograft+FK506 + GM6001 group were significantly lower than those of allograft+FK506 group (P < .05). MMP-8 and MMP-9 protein expression in the grafts of allograft+FK506 + GM6001 group were lower than those of allograft+FK506 group verified by immunohistochemical staining and western blotting. CONCLUSION FK506 combined with GM6001 could alleviate tracheal obliteration in mouse heterotopic tracheal transplantation model, due to its inhibitory effect on MMPs.
Collapse
Affiliation(s)
- Yiqian Li
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Ping Shu
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Liang Tang
- Department of central Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Xiaojun Yang
- Department of central Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Junwei Fan
- Department of general Surgery, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China.
| | - Xiaoqing Zhang
- Department of pharmacy, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China.
| |
Collapse
|
27
|
Thimraj TA, Sompa SI, Ganguly K, Ernstgård L, Johanson G, Palmberg L, Upadhyay S. Evaluation of diacetyl mediated pulmonary effects in physiologically relevant air-liquid interface models of human primary bronchial epithelial cells. Toxicol In Vitro 2019; 61:104617. [PMID: 31381966 DOI: 10.1016/j.tiv.2019.104617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/03/2019] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
Diacetyl is an artificial flavouring agent, known to cause bronchiolitis obliterans. Diacetyl-induced pulmonary effects were assessed in human primary bronchial epithelial cells (PBEC) cultured at air-liquid interface (ALI). The PBEC-ALI models were exposed to clean air (sham) and diacetyl vapour (1, 3, 10 and 30 ppm) for 30 min. At 6 and 24 h post-exposure, cell medium was sampled for assessment of cytotoxicity measurement, and CXCL8, MMP9 secretion by ELISA. Pro-inflammatory, oxidative stress, tissue injury/repair, anti-protease and beta-defensin markers were assessed using qRT-PCR. Additionally, epidermal growth factor receptor ligands (amphiregulin) and anti-protease (SLPI) were analysed at 6 h, 8 h and 24 h post exposure to 1 and 10 ppm diacetyl. No significant cytotoxicity was observed at any exposure level. MMP9 was significantly increased in both apical and basal media at 24 h. Both SLPI and amphiregulin secretion were significantly increased following exposure to 10 ppm diacetyl. Exposure of PBEC-ALI model to diacetyl vapour resulted in significantly altered transcript expression of pro-inflammatory, oxidative stress, anti-protease, tissue injury/repair markers. Changes in transcript expression of significantly altered markers were more prominent 24 h post-exposure compared to 6 h. This study warrants further mechanistic investigations to elucidate the pulmonary effects of inhaled diacetyl vapour using physiologically relevant in vitro models.
Collapse
Affiliation(s)
- Tania A Thimraj
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Shanzina I Sompa
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Koustav Ganguly
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Lena Ernstgård
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Gunnar Johanson
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Lena Palmberg
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Swapna Upadhyay
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Courtwright AM, Lamattina AM, Louis PH, Trindade AJ, Burkett P, Imani J, Shrestha S, Divo M, Keller S, Rosas IO, Goldberg HJ, El-Chemaly S. Hyaluronan and LYVE-1 and allograft function in lung transplantation recipients. Sci Rep 2019; 9:9003. [PMID: 31227795 PMCID: PMC6588572 DOI: 10.1038/s41598-019-45309-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/04/2019] [Indexed: 11/23/2022] Open
Abstract
Hyaluronan (HA) is associated with innate immune response activation and may be a marker of allograft dysfunction in lung transplant recipients. This was a prospective, single center study comparing levels of bronchioalveolar lavage (BAL) and serum HA and the HA immobilizer LYVE-1 in lung transplant recipients with and without acute cellular rejection (ACR). Chronic lung allograft dysfunction (CLAD)-free survival was also evaluated based on HA and LYVE-1 levels. 78 recipients were enrolled with a total of 115 diagnostic biopsies and 1.5 years of median follow-up. Serum HA was correlated with BAL HA (r = 0.25, p = 0.01) and with serum LYVE-1 (r = 0.32, p = 0.002). There was significant variation in HA and LYVE-1 over time, regardless of ACR status. Levels of serum HA (median 74.7 vs 82.7, p = 0.69), BAL HA (median 149.4 vs 134.5, p = 0.39), and LYVE-1 (mean 190.2 vs 183.8, p = 0.72) were not associated with ACR. CLAD-free survival was not different in recipients with any episode of elevated serum HA (HR = 1.5, 95% CI = 0.3–7.7, p = 0.61) or BAL HA (HR = 0.94, 95% CI = 0.2–3.6, p = 0.93). These results did not differ when stratified by bilateral transplant status. In this small cohort, serum HA, BAL HA, and LYVE-1 levels are not associated with ACR or CLAD-free survival in lung transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | - Jewel Imani
- Brigham and Women's Hospital, Boston, MA, United States
| | | | - Miguel Divo
- Brigham and Women's Hospital, Boston, MA, United States
| | - Steve Keller
- Brigham and Women's Hospital, Boston, MA, United States
| | - Ivan O Rosas
- Brigham and Women's Hospital, Boston, MA, United States
| | | | | |
Collapse
|
29
|
Abstract
INTRODUCTION Bronchiolitis obliterans (BO) is a chronic and irreversible obstructive lung disease leading to the obstruction and/or obliteration of the small airways. Three main BO entities are distinguished: post-infectious BO (PIBO); BO post lung transplantation; and BO after bone marrow transplantation (BMT) or hematopoietic stem cell transplantation (HSCT). All three entities are separate, however, there are similarities in histopathological characteristics and possibly in aspects of the development pathway. Areas covered: We review current evidence of bronchiolitis obliterans diagnosis and management in children. The diagnosis of BO is usually based on a combination of history, clinical and radiological findings, although lung biopsy and histopathology remain the gold standard approaches to confirm BO. Expert opinion: At present, we do not have a clear understanding of the mechanisms of the development of BO and lack strong evidence for treatment. Although most BO in children is post-infectious, most of the current evidence for treatment originates from studies analyzing BO in adult lung transplant and HSCT patients. BO management requires multidisciplinary approach and care in specialized centers.
Collapse
Affiliation(s)
- Ema Kavaliunaite
- a Respiratory Unit , Great Ormond Street Hospital for Children NHS Foundation Trust , London , UK
| | - Paul Aurora
- a Respiratory Unit , Great Ormond Street Hospital for Children NHS Foundation Trust , London , UK.,b Respiratory Critical Care and Anaesthesia Section , Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health , London , UK
| |
Collapse
|
30
|
Yang JYC, Verleden SE, Zarinsefat A, Vanaudenaerde BM, Vos R, Verleden GM, Sarwal RD, Sigdel TK, Liberto JM, Damm I, Watson D, Sarwal MM. Cell-Free DNA and CXCL10 Derived from Bronchoalveolar Lavage Predict Lung Transplant Survival. J Clin Med 2019; 8:jcm8020241. [PMID: 30781765 PMCID: PMC6406976 DOI: 10.3390/jcm8020241] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
Standard methods for detecting chronic lung allograft dysfunction (CLAD) and rejection have poor sensitivity and specificity and have conventionally required bronchoscopies and biopsies. Plasma cell-free DNA (cfDNA) has been shown to be increased in various types of allograft injury in transplant recipients and CXCL10 has been reported to be increased in the lung tissue of patients undergoing CLAD. This study used a novel cfDNA and CXCL10 assay to evaluate the noninvasive assessment of CLAD phenotype and prediction of survival from bronchoalveolar lavage (BAL) fluid. A total of 60 BAL samples (20 with bronchiolitis obliterans (BOS), 20 with restrictive allograft syndrome (RAS), and 20 with stable allografts (STA)) were collected from 60 unique lung transplant patients; cfDNA and CXCL10 were measured by the ELISA-based KIT assay. Median cfDNA was significantly higher in BOS patients (6739 genomic equivalents (GE)/mL) versus STA (2920 GE/mL) and RAS (4174 GE/mL) (p < 0.01 all comparisons). Likelihood ratio tests revealed a significant association of overall survival with cfDNA (p = 0.0083), CXCL10 (p = 0.0146), and the interaction of cfDNA and CXCL10 (p = 0.023) based on multivariate Cox proportional hazards regression. Dichotomizing patients based on the median cfDNA level controlled for the mean level of CXCL10 revealed an over two-fold longer median overall survival time in patients with low levels of cfDNA. The KIT assay could predict allograft survival with superior performance compared with traditional biomarkers. These data support the pursuit of larger prospective studies to evaluate the predictive performance of cfDNA and CXCL10 prior to lung allograft failure.
Collapse
Affiliation(s)
- Joshua Y C Yang
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
- KIT Bio, 2000 University Avenue, Palo Alto, CA 94303, USA.
| | - Stijn E Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium.
| | - Arya Zarinsefat
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium.
| | - Robin Vos
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium.
| | - Geert M Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium.
| | - Reuben D Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Tara K Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Juliane M Liberto
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Izabella Damm
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Drew Watson
- KIT Bio, 2000 University Avenue, Palo Alto, CA 94303, USA.
| | - Minnie M Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
- KIT Bio, 2000 University Avenue, Palo Alto, CA 94303, USA.
| |
Collapse
|
31
|
Sacreas A, Yang JYC, Vanaudenaerde BM, Sigdel TK, Liberto JM, Damm I, Verleden GM, Vos R, Verleden SE, Sarwal MM. The common rejection module in chronic rejection post lung transplantation. PLoS One 2018; 13:e0205107. [PMID: 30289917 PMCID: PMC6173434 DOI: 10.1371/journal.pone.0205107] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/19/2018] [Indexed: 11/19/2022] Open
Abstract
Rationale Recent studies suggest that similar injury mechanisms are in place across different solid organ transplants, resulting in the identification of a common rejection module (CRM), consisting of 11 genes that are overexpressed during acute and, to a lesser extent, chronic allograft rejection. Objectives We wanted to evaluate the usefulness of the CRM module in identifying acute rejection (AR) and different phenotypes of chronic lung transplant rejection (CLAD), i.e., bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS), using transbronchial brushings, broncho-alveolar lavage (BAL) samples, and explant tissue. Methods Gene expression measurements for the 11 CRM genes (CD6, TAP1, CXCL10, CXCL9, INPP5D, ISG20, LCK, NKG7, PSMB9, RUNX3, and BASP1) were performed via qRT-PCR in 14 transbronchial brushings (AR, n = 4; no AR, n = 10), 32 BAL samples (stable, n = 13; AR, n = 8; BOS, n = 9; RAS, n = 10), and 44 tissue specimens (unused donor lungs, n = 15; BOS, n = 13; RAS, n = 16). A geometric mean score was calculated to quantitate overall burden of immune injury and a new computational model was built for the most significant genes in lung transplant injury. Results Acute rejection showed a significant difference in almost every gene analysed, validating previous observations from microarray analysis. RAS tissue demonstrated a higher geometric mean score (6.35) compared to donor tissue (4.09, p = 0.018). Analysis of individual CRM genes showed an increased expression of ISG20, CXCL10 and CXCL9 in RAS. In BAL samples, no differences were detected in gene expression or geometric mean scores between the various groups (stable, 5.15; AR, 5.81; BOS, 5.62; RAS, 7.31). A newly modelled 2-gene tissue CRM score did not demonstrate any difference between BOS and RAS (p>0.05). However, the model was able to discriminate RAS from BOS tissue (AUC = 0.75, 95% CI = 0.55–0.94, p = 0.025). Conclusion Transcriptional tissue analysis for CRM genes in CLAD can identify acute rejection and distinguish RAS from BOS. The immune activation in RAS seems similar to acute rejection after kidney/liver/heart transplantation.
Collapse
Affiliation(s)
- Annelore Sacreas
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Joshua Y. C. Yang
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Bart M. Vanaudenaerde
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Tara K. Sigdel
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Juliane M. Liberto
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Izabella Damm
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
| | - Geert M. Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Stijn E. Verleden
- Leuven Lung Transplant Unit, Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- * E-mail: (SEV); (MMS)
| | - Minnie M. Sarwal
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail: (SEV); (MMS)
| |
Collapse
|
32
|
Lung Transplantation in Idiopathic Pulmonary Fibrosis. Med Sci (Basel) 2018; 6:medsci6030068. [PMID: 30142942 PMCID: PMC6164271 DOI: 10.3390/medsci6030068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Despite the advances in recent years in the treatment of idiopathic pulmonary fibrosis (IPF), it continues to be a progressive disease with poor prognosis. In selected patients, lung transplantation may be a treatment option, with optimal results in survival and quality of life. Currently, pulmonary fibrosis is the main cause of lung transplantation. However, mortality on the waiting list of these patients is high, since many patients are referred to the transplant units with advanced disease. There is not a parameter that can predict the survival of a specific patient. Different variables are to be considered in order to decide the right time to send them to a transplant unit. It is also very difficult to decide when to include these patients on the waiting list. Every patient diagnosed with IPF, without contraindications for surgery, should be referred early to a transplant unit for assessment. A uni or bilateral transplantation will be decided based on the characteristics of the patient and the experience of each center. The post-transplant survival of recipients with IPF is lower than that observed in other diseases, such as cystic fibrosis or chronic obstructive pulmonary disease as a consequence of their older age and the frequent presence of associated comorbidity. Post-transplant follow-up must be tight in order to assure optimal level of immunosuppressive treatment, detect complications associated with it, and avoid graft rejection. The main cause of long-term mortality is late graft dysfunction as a consequence of chronic rejection. Other complications, such as infections and tumors, must be considered.
Collapse
|
33
|
Gwinn WM, Flake GP, Bousquet RW, Taylor GJ, Morgan DL. Airway injury in an in vitro human epithelium-fibroblast model of diacetyl vapor exposure: diacetyl-induced basal/suprabasal spongiosis. Inhal Toxicol 2018; 29:310-321. [PMID: 28984536 DOI: 10.1080/08958378.2017.1369604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inhalation exposure to diacetyl (DA) is associated with obliterative bronchiolitis (OB) in workers and induces OB-like fibrotic airway lesions in rats. The pathogenesis of OB is poorly understood in part due to complex interactions between airway epithelial, mesenchymal and blood-derived inflammatory cells. DA-induced airway toxicity in the absence of recruited-inflammatory/immune cells was characterized using an air-liquid interface (ALI) model consisting of human airway epithelium with (Epi/FT) and without (Epi) a mesenchymal component. ALI cultures were exposed to 25 mM DA-derived vapors (using vapor cups) for 1 h on day 0, 2 and 4. In some experiments, the tissues were exposed to 2,3-hexanedione (Hex) which is structurally-similar, but much less fibrogenic than DA. Lactate dehydrogenase activity and day 6 histopathologic changes associated with epithelial injury, including basal/suprabasal spongiosis, were increased following exposure of Epi/FT tissues to DA but not control or Hex vapors. IL-1a, IL-6, IL-8, sIL-1Ra, TGFa, MCP-3 and TNFa proteins were increased following DA exposure of Epi/FT tissues; only IL-1a, IL-8, sIL-1Ra and TGFa were increased following exposure of Epi tissues. MMP-1, MMP-3 and TIMP-1 proteins were increased following DA exposure of Epi/FT tissues; whereas MMP-2, MMP-7 and TIMP-2 were decreased, and production was largely dependent upon the presence of sub-epithelial stromal matrix/fibroblasts. Hex-induced protein changes were minimal. This in vitro study demonstrated that exposure of human airways to DA vapors induced epithelial injury (with the histopathologic feature of basal/suprabasal spongiosis) and increased release of pro-inflammatory and pro-fibrotic cytokines/chemokines as well as MMPs/TIMPs in the absence of recruited-inflammatory cells.
Collapse
Affiliation(s)
- William M Gwinn
- a NTP Laboratory, Division of the National Toxicology Program (DNTP) , National Institute of Environmental Health Sciences (NIEHS) , Durham , NC , USA
| | - Gordon P Flake
- b Cell and Molecular Pathology Branch, DNTP, NIEHS , Durham , NC , USA
| | - Ronald W Bousquet
- c Alion Science and Technology Corporation , Research Triangle Park, Durham , NC , USA
| | - Genie J Taylor
- c Alion Science and Technology Corporation , Research Triangle Park, Durham , NC , USA
| | - Daniel L Morgan
- a NTP Laboratory, Division of the National Toxicology Program (DNTP) , National Institute of Environmental Health Sciences (NIEHS) , Durham , NC , USA
| |
Collapse
|
34
|
Upregulation of alveolar neutrophil enzymes and long pentraxin-3 in human chronic lung allograft dysfunction subtypes. J Thorac Cardiovasc Surg 2018; 155:2774-2776.e2. [DOI: 10.1016/j.jtcvs.2018.02.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/17/2018] [Indexed: 11/23/2022]
|
35
|
Sequential broncho-alveolar lavages reflect distinct pulmonary compartments: clinical and research implications in lung transplantation. Respir Res 2018; 19:102. [PMID: 29801490 PMCID: PMC5970521 DOI: 10.1186/s12931-018-0786-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/20/2018] [Indexed: 11/10/2022] Open
Abstract
Background Bronchoalveolar lavage (BAL) has proven to be very useful to monitor the lung allograft after transplantation. In addition to allowing detection of infections, multiple BAL analytes have been proposed as potential biomarkers of lung allograft rejection or dysfunction. However, BAL collection is not well standardized and differences in BAL collection represent an important source of variation. We hypothesized that there are systematic differences between sequential BALs that are relevant to BAL analysis. Methods As part of 126 consecutive bronchoscopies in lung transplant recipients, two sequential BALs (BAL1 and BAL2) were performed in one location during each bronchoscopy by instilling and suctioning 50 ml of normal saline twice into separate containers. Cell concentration, viability and differentials, Surfactant Protein-D (SP-D), Club Cell Secretory Protein (CCSP), and levels of CXCL10, IL-10, CCL2, CCL5, VEGF-C, RAGE, CXCL9, CXCL1, IL-17A, IL-21, PDGF, and GCSF were compared between BAL1 and BAL2. Results Total cell concentration did not differ between BAL1 and BAL2; however, compared to BAL2, BAL1 had more dead cells, epithelial cells, neutrophils, and higher concentrations of airway epithelium-derived CCSP and inflammatory markers. BAL2 had a higher concentration of SP-D compared to BAL1. Conclusion In this study performed in lung transplant recipients, we show that sequential BALs represent different lung compartments and have distinct compositions. BAL1 represents the airway compartment with more epithelial cells, neutrophils, and epithelium-derived CCSP. Conversely, BAL2 samples preferentially the distal bronchoalveolar space with greater cell viability and higher SP-D. Our findings illustrate how the method of BAL collection can influence analyte concentrations and further emphasize the need for a standardized approach in translational research involving BAL samples. Electronic supplementary material The online version of this article (10.1186/s12931-018-0786-z) contains supplementary material, which is available to authorized users.
Collapse
|
36
|
Schott C, Weigt SS, Turturice BA, Metwally A, Belperio J, Finn PW, Perkins DL. Bronchiolitis obliterans syndrome susceptibility and the pulmonary microbiome. J Heart Lung Transplant 2018; 37:1131-1140. [PMID: 29929823 DOI: 10.1016/j.healun.2018.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/30/2018] [Accepted: 04/18/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Lung transplantation outcomes remain complicated by bronchiolitis obliterans syndrome (BOS), a major cause of mortality and retransplantation for patients. A variety of factors linking inflammation and BOS have emerged, meriting further exploration of the microbiome as a source of inflammation. In this analysis, we determined features of the pulmonary microbiome associated with BOS susceptibility. METHODS Bronchoalveolar lavage (BAL) samples were collected from 25 patients during standard of care bronchoscopies before BOS onset. Microbial DNA was isolated from BAL fluid and prepared for metagenomics shotgun sequencing. Patient microbiomes were phenotyped using k-means clustering and compared to determine effects on BOS-free survival. RESULTS Clustering identified 3 microbiome phenotypes: Actinobacteria dominant (AD), mixed, and Proteobacteria dominant. AD microbiomes, distinguished by enrichment with Gram-positive organisms, conferred reduced odds and risks for patients to develop acute rejection and BOS compared with non-AD microbiomes. These findings were independent of treatment models. Microbiome findings were correlated with BAL cell counts and polymorphonuclear cell percentages. CONCLUSIONS In some populations, features of the microbiome may be used to assess BOS susceptibility. Namely, a Gram-positive enriched pulmonary microbiome may predict resilience to BOS.
Collapse
Affiliation(s)
- Cody Schott
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - S Samuel Weigt
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California at Los Angeles, Los Angeles, California
| | - Benjamin A Turturice
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Ahmed Metwally
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - John Belperio
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California at Los Angeles, Los Angeles, California
| | - Patricia W Finn
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - David L Perkins
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Surgery, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
37
|
Yin J, Ma X, Huang F, Ma Y, Li Y. Bronchiolitis obliterans murine model induced by nitric acid aerosol inhalation: An economical and reproducible model. Exp Lung Res 2018; 44:143-152. [PMID: 29683731 DOI: 10.1080/01902148.2018.1455926] [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] [Indexed: 10/17/2022]
Abstract
PURPOSE Bronchiolitis obliterans (BO) is a highly debilitative and fatal syndrome associated with a series of severe lower airway disorders. The pathogenesis of BO is complicated and not entirely understood. An appropriate animal model of BO may aid research into its pathogenesis. Here, we establish a mouse model of BO to provide insight into this disease. MATERIALS AND METHODS 6-8 week old BABL/c mice were exposed to 5% nitric acid (NA) aerosol through a nebulizer for 3 hours, and controls were exposed to distilled water instead. Symptoms, airway resistance and pathological process were observed dynamically. The levels of matrix metalloproteinase-2 (MMP-2), MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), 8-isoprostane and myeloperoxidase (MPO) in lung tissue and bronchoalveolar lavage fluids (BLAF) were determined by ELISA on day 3, 7, 14, 28 and 56 after the aerosol nebulization. RESULTS Typical BO lesions were observed in NA nebulized mice characterized histologically by initial necrotizing bronchiolitis and final airway fibrosis at day 28 after the aerosol nebulization. NA nebulized mice also exhibited labored breathing and significantly increased airway resistance. Expression of MMP-2, MMP-9, TIMP-1, 8-isoprostane and MPO were significantly elevated in NA nebulized mice in different time frame. CONCLUSION A murine BO model was established by NA aerosol inhalation. It provides an easy, economic, and reproducible mice model for BO research.
Collapse
Affiliation(s)
- Jianing Yin
- a Department of Pediatrics , The First Hospital of Jilin University , Changchun , jilin , China.,b Department of Molecular Biology , Basic Medical College of Jilin University , Changchun , jilin , China
| | - Xiaobo Ma
- c Department of Pathology , The First Hospital of Jilin University , Changchun , jilin , China
| | - Fei Huang
- d Department of Orthopedics , China-Japan Union Hospital of Jilin University , Changchun , jilin , China
| | - Yucong Ma
- a Department of Pediatrics , The First Hospital of Jilin University , Changchun , jilin , China
| | - Yanan Li
- a Department of Pediatrics , The First Hospital of Jilin University , Changchun , jilin , China.,b Department of Molecular Biology , Basic Medical College of Jilin University , Changchun , jilin , China
| |
Collapse
|
38
|
Inhibition of T Cell Alloreactivity by Bronchial Epithelium Is Impaired in Lung Transplant Recipients, Through Pathways Involving TGF-β, IL-10 and HLA-G. Transplantation 2017; 101:2192-2199. [PMID: 27820781 DOI: 10.1097/tp.0000000000001553] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) after lung transplantation (LTx) results from bronchial epithelial cell (BECs) damages, thought to be orchestrated by T cells primed by antigen-presenting cell presenting alloantigens. In this cell cross-talk, BECs are also suspected to play a pivotal immunosuppressive role in T cell alloreactivity. We studied the immunomodulating role of BECs in a human ex vivo model of allogeneic T cell response, both in healthy subjects and LTx recipients. METHODS BECs from 35 LTx recipients (n = 22 stable, n = 13 BOS) and healthy controls (n = 25) were cultured as primary cell cultures. Their inhibitory capacities through the involvement of tolerogenic molecules (HLA-G, TGF-β, and IL-10) were tested on a mixed lymphocyte reaction between antigen-presenting cells and recipient T cells. RESULTS Control BECs inhibited T cell alloproliferation by a mean of 53 ± 7%. This inhibitory effect of BECs was significantly reduced in the stable LTx group (24 ± 8%, P = 0.009), but not in the BOS TxP group (53 ± 10%, P = 0.97). Neutralization of HLA-G, TGF-β, and IL-10 partially restored T cell alloproliferation, arguing for their involvement in the immunosuppressive effect of BECs. BECs culture supernatant from stable LTx patients with impaired BEC properties showed a skewed Th2-type secretion profile (high IL-4/IFN-γ ratio). CONCLUSIONS The inhibitory properties of BECs are dysregulated in stable LTx recipients, which could suggest their instrumental role in the initiation of BOS process and potential targeted therapies.
Collapse
|
39
|
Abstract
Chronic lung allograft dysfunction (CLAD) is the major limitation to posttransplant survival. This review highlights the evolving definition of CLAD, risk factors, treatment, and expected outcomes after the development of CLAD.
Collapse
|
40
|
Verleden SE, Vos R, Vanaudenaerde BM, Verleden GM. Chronic lung allograft dysfunction phenotypes and treatment. J Thorac Dis 2017; 9:2650-2659. [PMID: 28932572 DOI: 10.21037/jtd.2017.07.81] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle limiting long-term survival post lung transplantation. Given the clinical heterogeneity of CLAD, recently two phenotypes of CLAD have been defined [bronchiolitis obliterans syndrome (BOS) vs. restrictive allograft syndrome (RAS) or restrictive CLAD (rCLAD)]. BOS is characterized by an obstructive pulmonary function, air trapping on CT and obliterative bronchiolitis (OB) on histopathology, while RAS/rCLAD patients show a restrictive pulmonary function, persistent pleuro-parenchymal infiltrates on CT and pleuroparenchymal fibro-elastosis on biopsies. Importantly, the patients with RAS/rCLAD have a severely limited survival post diagnosis of 6-18 months compared to 3-5 years after BOS diagnosis. In this review, we will review historical evidence for this heterogeneity and we will highlight the clinical, radiological, histopathological characteristics of both phenotypes, as well as their risk factors. Treatment of CLAD remains troublesome, nevertheless, we will give an overview of different treatment strategies that have been tried with some success. Adequate phenotyping remains difficult but is clearly needed for both clinical and scientific purposes.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Brass DM, Gwinn WM, Valente AM, Kelly FL, Brinkley CD, Nagler AE, Moseley MA, Morgan DL, Palmer SM, Foster MW. The Diacetyl-Exposed Human Airway Epithelial Secretome: New Insights into Flavoring-Induced Airways Disease. Am J Respir Cell Mol Biol 2017; 56:784-795. [PMID: 28248570 DOI: 10.1165/rcmb.2016-0372oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bronchiolitis obliterans (BO) is an increasingly important lung disease characterized by fibroproliferative airway lesions and decrements in lung function. Occupational exposure to the artificial food flavoring ingredient diacetyl, commonly used to impart a buttery flavor to microwave popcorn, has been associated with BO development. In the occupational setting, diacetyl vapor is first encountered by the airway epithelium. To better understand the effects of diacetyl vapor on the airway epithelium, we used an unbiased proteomic approach to characterize both the apical and basolateral secretomes of air-liquid interface cultures of primary human airway epithelial cells from four unique donors after exposure to an occupationally relevant concentration (∼1,100 ppm) of diacetyl vapor or phosphate-buffered saline as a control on alternating days. Basolateral and apical supernatants collected 48 h after the third exposure were analyzed using one-dimensional liquid chromatography tandem mass spectrometry. Paired t tests adjusted for multiple comparisons were used to assess differential expression between diacetyl and phosphate-buffered saline exposure. Of the significantly differentially expressed proteins identified, 61 were unique to the apical secretome, 81 were unique to the basolateral secretome, and 11 were present in both. Pathway enrichment analysis using publicly available databases revealed that proteins associated with matrix remodeling, including degradation, assembly, and new matrix organization, were overrepresented in the data sets. Similarly, protein modifiers of epidermal growth factor receptor signaling were significantly altered. The ordered changes in protein expression suggest that the airway epithelial response to diacetyl may contribute to BO pathogenesis.
Collapse
Affiliation(s)
- David M Brass
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine
| | - William M Gwinn
- 2 National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | | - Andrew E Nagler
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine
| | - M Arthur Moseley
- 4 Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, North Carolina; and
| | - Daniel L Morgan
- 2 National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Scott M Palmer
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine
| | - Matthew W Foster
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine.,4 Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, North Carolina; and
| |
Collapse
|
42
|
Dilek F, Emin Ö, Gültepe B, Yazıcı M, Çakır E, Gedik AH. Evaluation of nasal fluid β-defensin 2 levels in children with allergic rhinitis. Turk Arch Pediatr 2017; 52:79-84. [PMID: 28747838 DOI: 10.5152/turkpediatriars.2017.4497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 02/22/2016] [Indexed: 01/14/2023]
Abstract
AIM Knowledge about the role of the innate immune system in the pathogenesis of allergic diseases has been expanding in recent years. Defensins are antimicrobial peptides that are components of the innate immune system. Defensins have strong efficacy against bacterial, viral, and fungal infections. Moreover, they have regulatory functions in many physiologic processes such as antitumoral immunity, chemotaxis, inflammation, and wound healing. In this study, we aimed to investigate β-defensin 2 levels in the nasal fluids of children with allergic rhinitis. MATERIAL AND METHODS Study and control groups consisted of 28 patients with newly diagnosed allergic rhinitis who were not taking any medication, and 23 healthy children. Skin prick tests were performed on patients with allergic rhinitis and disease severity was assessed using the total symptom score. Nasal fluid samples were obtained using a modified polyurethane sponge absorption method from patients and control subjects. Nasal fluid β-defensin 2 levels were determined using an enzyme-linked immunosorbent assay (ELISA). RESULTS The median value of nasal fluid β-defensin 2 levels were 173.8 pg/mL (interquartile range; 54.8-205.9 pg/mL) in allergic rhinitis group and 241.6 pg/mL (163.5-315.2 pg/mL) in the control group. There was a statistically significant difference between the two groups (p=0.01). Moreover, nasal fluid β-defensin 2 levels showed a significant negative correlation with total symptom scores (rho= -0.78, p<0.001). CONCLUSIONS Children with allergic rhinitis have reduced nasal fluid β-defensin 2 levels compared with controls, and β-defensin 2 levels were negatively correlated with disease severity. A more definite understanding of the roles of defensins and other antimicrobial peptides in allergic inflammation can open up new horizons in the management and treatment of these common diseases.
Collapse
Affiliation(s)
- Fatih Dilek
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| | - Özkaya Emin
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| | - Bilge Gültepe
- Department of Clinical Microbiology, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| | - Mebrure Yazıcı
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| | - Erkan Çakır
- Department of Pediatrics, Division of Pediatric Chest Diseases, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| | - Ahmet Hakan Gedik
- Department of Pediatrics, Division of Pediatric Chest Diseases, Bezmialem Vakif University School of Medicine, İstanbul, Turkey
| |
Collapse
|
43
|
Pain M, Royer PJ, Loy J, Girardeau A, Tissot A, Lacoste P, Roux A, Reynaud-Gaubert M, Kessler R, Mussot S, Dromer C, Brugière O, Mornex JF, Guillemain R, Dahan M, Knoop C, Botturi K, Pison C, Danger R, Brouard S, Magnan A. T Cells Promote Bronchial Epithelial Cell Secretion of Matrix Metalloproteinase-9 via a C-C Chemokine Receptor Type 2 Pathway: Implications for Chronic Lung Allograft Dysfunction. Am J Transplant 2017; 17:1502-1514. [PMID: 27982503 DOI: 10.1111/ajt.14166] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/30/2016] [Accepted: 12/04/2016] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major limitation of long-term survival after lung transplantation. CLAD manifests as bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS). Alloimmune reactions and epithelial-to-mesenchymal transition have been suggested in BOS. However, little is known regarding the role of allogenicity in epithelial cell differentiation. Primary human bronchial epithelial cells (BECs) were treated with activated T cells in the presence or absence of transforming growth factor (TGF)-β. The expression of epithelial and mesenchymal markers was investigated. The secretion of inflammatory cytokines and matrix metalloproteinase (MMP)-9 was measured in culture supernatants and in plasma from lung transplant recipients (LTRs): 49 stable, 29 with BOS, and 16 with RAS. We demonstrated that C-C motif chemokine 2 secreted by T cells supports TGF-β-induced MMP-9 production by BECs after binding to C-C chemokine receptor type 2. Longitudinal investigation in LTRs revealed a rise in plasma MMP-9 before CLAD onset. Multivariate analysis showed that plasma MMP-9 was independently associated with BOS (odds ratio [OR] = 6.19, p = 0.002) or RAS (OR = 3.9, p = 0.024) and predicted the occurrence of CLAD 12 months before the functional diagnosis. Thus, immune cells support airway remodeling through the production of MMP-9. Plasma MMP-9 is a potential predictive biomarker of CLAD.
Collapse
Affiliation(s)
- M Pain
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P-J Royer
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - J Loy
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Girardeau
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Tissot
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P Lacoste
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Roux
- Hôpital Foch, Suresnes, Université Versailles Saint-Quentin-en-Yvelines, UPRES EA220, Versailles, France
| | | | - R Kessler
- CHU de Strasbourg, Strasbourg, France
| | - S Mussot
- Centre Chirurgical Marie Lannelongue, Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardiopulmonaire, Le Plessis Robinson, France
| | - C Dromer
- CHU de Bordeaux, Bordeaux, France
| | - O Brugière
- Hôpital Bichat, Service de Pneumologie et Transplantation Pulmonaire, Paris, France
| | - J-F Mornex
- Université de Lyon, INRA, UMR754, Lyon, Hospices Civils de Lyon, Lyon, France
| | | | - M Dahan
- CHU de Toulouse, Toulouse, France
| | - C Knoop
- Hôpital Erasme, Bruxelles, Belgique
| | - K Botturi
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - C Pison
- Clinique Universitaire Pneumologie, Pôle Thorax et Vaisseaux, CHU de Grenoble, Université de Grenoble, INSERM U1055, Grenoble, France
| | - R Danger
- Université de Nantes, INSERM U1064 and Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - S Brouard
- Université de Nantes, INSERM U1064 and Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - A Magnan
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | | |
Collapse
|
44
|
Costa J, Benvenuto LJ, Sonett JR. Long-term outcomes and management of lung transplant recipients. Best Pract Res Clin Anaesthesiol 2017; 31:285-297. [PMID: 29110800 DOI: 10.1016/j.bpa.2017.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
Lung transplantation is an established treatment for patients with end-stage lung disease. Improvements in immunosuppression and therapeutic management of infections have resulted in improved long-term survival and a decline in allograft rejection. Allograft rejection continues to be a serious complication following lung transplantation, thereby leading to acute graft failure and, subsequently, chronic lung allograft dysfunction (CLAD). Bronchiolitis obliterans syndrome (BOS), the most common phenotype of CLAD, is the leading cause of late mortality and morbidity in lung recipients, with 50% having developed BOS within 5 years of lung transplantation. Infections in lung transplant recipients are also a significant complication and represent the most common cause of death within the first year. The success of lung transplantation depends on careful management of immunosuppressive regimens to reduce the rate of rejection, while monitoring recipients for infections and complications to help identify problems early. The long-term outcomes and management of lung transplant recipients are critically based on modulating natural immune response of the recipient to prevent acute and chronic rejection. Understanding the immune mechanisms and temporal correlation of acute and chronic rejection is thus critical in the long-term management of lung recipients.
Collapse
Affiliation(s)
- Joseph Costa
- Columbia University College of Physicians and Surgeons, Columbia University Medical Center, 622 West 168th St, PH 14, Room 108, New York, NY 10032, USA.
| | - Luke J Benvenuto
- Columbia University College of Physicians and Surgeons, Division Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, 622 West 168th St, PH 14, Room 104, New York, NY 10032, USA.
| | - Joshua R Sonett
- The Price Family Center for Comprehensive Chest Care, Columbia University College of Physicians and Surgeons, Columbia University Medical Center, 161 Fort Washington Avenue, New York, NY 10032, USA.
| |
Collapse
|
45
|
Müller C, Andersson-Sjöland A, Schultz HH, Eriksson LT, Andersen CB, Iversen M, Westergren-Thorsson G. Early extracellular matrix changes are associated with later development of bronchiolitis obliterans syndrome after lung transplantation. BMJ Open Respir Res 2017; 4:e000177. [PMID: 28469930 PMCID: PMC5411729 DOI: 10.1136/bmjresp-2016-000177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022] Open
Abstract
Background Chronic lung allograft dysfunction in the form of bronchiolitis obliterans syndrome (BOS) is the main cause of death beyond 1-year post-lung transplantation. The disease-initiating triggers as well as the molecular changes leading to fibrotic alterations in the transplanted lung are largely unknown. The aim of this study was to identify potential early changes in the extracellular matrix (ECM) in different compartments of the transplanted lung prior to the development of BOS. Methods Transbronchial biopsies from a cohort of 58 lung transplantation patients at the Copenhagen University hospital between 2005 and 2006, with or without development of BOS in a 5-year follow-up, were obtained 3 and 12 months after transplantation. Biopsies were assessed for total collagen, collagen type IV and biglycan in the alveolar and small airway compartments using Masson's Trichrome staining and immunohistochemistry. Results A time-specific and compartment-specific pattern of ECM changes was detected. Alveolar total collagen (p=0.0190) and small airway biglycan (p=0.0199) increased between 3 and 12 months after transplantation in patients developing BOS, while collagen type IV (p=0.0124) increased in patients without BOS. Patients with early-onset BOS mirrored this increase. Patients developing grade 3 BOS showed distinct ECM changes already at 3 months. Patients with BOS with treated acute rejections displayed reduced alveolar total collagen (p=0.0501) and small airway biglycan (p=0.0485) at 3 months. Conclusions Patients with future BOS displayed distinct ECM changes compared with patients without BOS. Our data indicate an involvement of alveolar and small airway compartments in post-transplantation changes in the development of BOS.
Collapse
Affiliation(s)
- Catharina Müller
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Hans Henrik Schultz
- Section for Lung Transplantation, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Leif T Eriksson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Claus B Andersen
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Martin Iversen
- Section for Lung Transplantation, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
46
|
Grønningsæter IS, Tsykunova G, Lilleeng K, Ahmed AB, Bruserud Ø, Reikvam H. Bronchiolitis obliterans syndrome in adults after allogeneic stem cell transplantation-pathophysiology, diagnostics and treatment. Expert Rev Clin Immunol 2017; 13:553-569. [DOI: 10.1080/1744666x.2017.1279053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ida Sofie Grønningsæter
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Galina Tsykunova
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Kyrre Lilleeng
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Aymen Bushra Ahmed
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Department of Medicine, Hematology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | |
Collapse
|
47
|
Im A, Hakim FT, Pavletic SZ. Novel targets in the treatment of chronic graft-versus-host disease. Leukemia 2016; 31:543-554. [PMID: 27899803 DOI: 10.1038/leu.2016.367] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
Abstract
Despite advances that have improved survival after allogeneic hematopoietic stem cell transplantation (HCT), chronic graft-versus-host disease (GVHD) remains a leading cause of late morbidity and mortality after transplant. Current treatment options show limited efficacy in steroid-refractory disease, and there exists a paucity of robust data to guide management decisions. Lack of United States Food and Drug Administration (FDA)- or European Medicines Agency (EMA)-approved agents in GVHD underscore the importance of developing novel therapies. Better understanding of the biology of chronic GVHD has provided novel targets for treatment, and structured guidelines in diagnosis and in clinical trial design have provided a common language and pathways for research in this area. These, combined with the surge of drug development in Oncology and Immunology, are factors that have contributed to the accelerating field of drug development and clinical research in chronic GVHD. In these exciting times, it is possible to foresee long awaited advances in the treatment of this devastating complication of HCT. This review will summarize the ongoing clinical development for novel therapies in chronic GVHD.
Collapse
Affiliation(s)
- A Im
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Cancer Institute and UPMC Cancer Centers, Pittsburgh, PA, USA.,Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - F T Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - S Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
|
49
|
Izykowski N, Kuehnel M, Hussein K, Mitschke K, Gunn M, Janciauskiene S, Haverich A, Warnecke G, Laenger F, Maus U, Jonigk D. Organizing pneumonia in mice and men. J Transl Med 2016; 14:169. [PMID: 27282780 PMCID: PMC4901413 DOI: 10.1186/s12967-016-0933-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/01/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Organizing pneumonia is a reaction pattern and an inflammatory response to acute lung injuries, and is characterized by intraluminal plugs of granulation tissue in distal airspaces. In contrast to other fibrotic pulmonary diseases, organizing pneumonia is generally responsive to corticosteroids. However, some patients do not respond to treatment, leading to respiratory failure and potentially death (up to 15 % of patients). In order to devise new therapeutic strategies, a better understanding of the disease's pathomechanisms is warranted. We previously generated a mouse model overexpressing CCL2, which generates organizing pneumonia-like changes, morphologically comparable to human patients. In this study, we investigated whether the histopathological similarities of human and murine pulmonary organizing pneumonia lesions also involve similar molecular pathways. METHODS We analyzed the similarities and differences of fibrosis-associated gene expression in individual compartments from patients with organizing pneumonia and transgenic (CCL2) mice using laser-assisted microdissection, real-time PCR and immunohistochemistry. RESULTS Gene expression profiling of human and murine organizing pneumonia lesions showed in part comparable expression levels of pivotal genes, notably of TGFB1/Tgfb1, TIMP1/Timp1, TIMP2/Timp2, COL3A1/Col3a1, CXCL12/Cxcl12, MMP2/Mmp2 and IL6/Il6. Hence, the transgenic CCL2 mouse model shows not only pathogenomic and morphological features of human organizing pneumonia but also a similar inflammatory profile. CONCLUSIONS We suggest that the CCL2-overexpressing transgenic mouse model (CCL2 Tg mice) is suitable for further investigation of fibrotic pulmonary remodeling, particularly of organizing pneumonia pathogenesis and for the search for novel therapeutic strategies.
Collapse
Affiliation(s)
- Nicole Izykowski
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany. .,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany.
| | - Mark Kuehnel
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Kais Hussein
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Kristin Mitschke
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Michael Gunn
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Sabina Janciauskiene
- Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Axel Haverich
- Department of Thoracic Surgery, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Gregor Warnecke
- Department of Thoracic Surgery, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Florian Laenger
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Ulrich Maus
- Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany.,German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Bad Nauheim, Germany
| |
Collapse
|
50
|
Verleden SE, Sacreas A, Vos R, Vanaudenaerde BM, Verleden GM. Advances in Understanding Bronchiolitis Obliterans After Lung Transplantation. Chest 2016; 150:219-25. [PMID: 27212132 DOI: 10.1016/j.chest.2016.04.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 03/29/2016] [Accepted: 04/13/2016] [Indexed: 11/30/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) remains a major complication after lung transplantation, causing significant morbidity and mortality in a majority of recipients. BOS is believed to be the clinical correlate of chronic allograft dysfunction, and is defined as an obstructive pulmonary function defect in the absence of other identifiable causes, mostly not amenable to treatment. Recently, it has become clear that BOS is not the only form of chronic allograft dysfunction and that other clinical phenotypes exist; however, we focus exclusively on BOS. Radiologic findings typically demonstrate air trapping, mosaic attenuation, and hyperinflation. Pathologic examination reveals obliterative bronchiolitis lesions and a pure obliteration of the small airways (< 2 mm), with a relatively normal surrounding parenchyma. In this review, we highlight recent advances in diagnosis, pathologic examination, and risk factors, such as microbes, viruses, and antibodies. Although the pathophysiological mechanisms remain largely unknown, we review the role of the airway epithelium and inflammation and the various experimental animal models. We also clarify the clinical and therapeutic implications of these findings. Although significant progress has been made, the exact pathophysiological mechanisms and adequate therapy for posttransplantation BOS remain unknown, highlighting the need for further research to improve long-term posttransplantation BOS-free and overall survival.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|