51
|
Wilkey BJ, Abrams BA. Mitigation of Primary Graft Dysfunction in Lung Transplantation: Current Understanding and Hopes for the Future. Semin Cardiothorac Vasc Anesth 2019; 24:54-66. [DOI: 10.1177/1089253219881980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Primary graft dysfunction (PGD) is a form of acute lung injury that develops within the first 72 hours after lung transplantation. The overall incidence of PGD is estimated to be around 30%, and the 30-day mortality for grade 3 PGD around 36%. PGD is also associated with the development of bronchiolitis obliterans syndrome, a specific form of chronic lung allograft dysfunction. In this article, we will discuss perioperative strategies for PGD prevention as well as possible future avenues for prevention and treatment.
Collapse
|
52
|
Parulekar AD, Kao CC. Detection, classification, and management of rejection after lung transplantation. J Thorac Dis 2019; 11:S1732-S1739. [PMID: 31632750 DOI: 10.21037/jtd.2019.03.83] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rejection is a major complication following lung transplantation. Acute cellular rejection, lymphocytic bronchiolitis, and antibody-mediated rejection (AMR) are all risk factors for the subsequent development of chronic lung allograft dysfunction (CLAD). Acute cellular rejection and lymphocytic bronchiolitis have well defined histopathologic diagnostic criteria and grading. Diagnosis of AMR requires a multidisciplinary approach. CLAD is the major barrier to long-term survival following lung transplantation. The most common phenotype of CLAD is bronchiolitis obliterans syndrome (BOS) which is defined by a persistent obstructive decline in lung function. Restrictive allograft dysfunction (RAS) is a second phenotype of CLAD and is associated with a worse prognosis. This article will review the diagnosis, staging, clinical presentation, and treatment of acute rejection, AMR, and CLAD following lung transplantation.
Collapse
Affiliation(s)
- Amit D Parulekar
- Section of Pulmonary, Critical Care, and Sleep, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christina C Kao
- Section of Pulmonary, Critical Care, and Sleep, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
53
|
Scozzi D, Ibrahim M, Liao F, Lin X, Hsiao HM, Hachem R, Tague LK, Ricci A, Kulkarni HS, Huang HJ, Sugimoto S, Krupnick AS, Kreisel D, Gelman AE. Mitochondrial damage-associated molecular patterns released by lung transplants are associated with primary graft dysfunction. Am J Transplant 2019; 19:1464-1477. [PMID: 30582269 PMCID: PMC6482093 DOI: 10.1111/ajt.15232] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/12/2018] [Accepted: 12/08/2018] [Indexed: 02/07/2023]
Abstract
Primary graft dysfunction (PGD) is a major limitation in short- and long-term lung transplant survival. Recent work has shown that mitochondrial damage-associated molecular patterns (mtDAMPs) can promote solid organ injury, but whether they contribute to PGD severity remains unclear. We quantitated circulating plasma mitochondrial DNA (mtDNA) in 62 patients, before lung transplantation and shortly after arrival to the intensive care unit. Although all recipients released mtDNA, high levels were associated with severe PGD development. In a mouse orthotopic lung transplant model of PGD, we detected airway cell-free damaged mitochondria and mtDNA in the peripheral circulation. Pharmacologic inhibition or genetic deletion of formylated peptide receptor 1 (FPR1), a chemotaxis sensor for N-formylated peptides released by damaged mitochondria, inhibited graft injury. An analysis of intragraft neutrophil-trafficking patterns reveals that FPR1 enhances neutrophil transepithelial migration and retention within airways but does not control extravasation. Using donor lungs that express a mitochondria-targeted reporter protein, we also show that FPR1-mediated neutrophil trafficking is coupled with the engulfment of damaged mitochondria, which in turn triggers reactive oxygen species (ROS)-induced pulmonary edema. Therefore, our data demonstrate an association between mtDAMP release and PGD development and suggest that neutrophil trafficking and effector responses to damaged mitochondria are drivers of graft damage.
Collapse
Affiliation(s)
- Davide Scozzi
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mohsen Ibrahim
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Fuyi Liao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Xue Lin
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Hsi-Min Hsiao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Ramsey Hachem
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Laneshia K Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Alberto Ricci
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Hrishikesh S Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Howard J Huang
- Houston Methodist J. C. Walter Jr. Transplant Center, Houston, Texas
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery, Okayama University Hospital, Okayama, Japan
| | - Alexander S Krupnick
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Daniel Kreisel
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew E Gelman
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
54
|
Bellier J, Lhommet P, Bonnette P, Puyo P, Le Guen M, Roux A, Parquin F, Chapelier A, Sage E. Extracorporeal membrane oxygenation for grade 3 primary graft dysfunction after lung transplantation: Long‐term outcomes. Clin Transplant 2019; 33:e13480. [DOI: 10.1111/ctr.13480] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 11/27/2022]
Affiliation(s)
| | - Pierre Lhommet
- Thoracic Surgery Department Foch Hospital Suresnes France
| | | | - Philippe Puyo
- Thoracic Surgery Department Foch Hospital Suresnes France
| | | | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Foch Hospital Suresnes France
| | | | | | - Edouard Sage
- Thoracic Surgery Department Foch Hospital Suresnes France
| | | |
Collapse
|
55
|
Weigt SS, Wang X, Palchevskiy V, Patel N, Derhovanessian A, Shino MY, Sayah DM, Lynch JP, Saggar R, Ross DJ, Kubak BM, Ardehali A, Palmer S, Husain S, Belperio JA. Gene Expression Profiling of Bronchoalveolar Lavage Cells During Aspergillus Colonization of the Lung Allograft. Transplantation 2019; 102:986-993. [PMID: 29256975 DOI: 10.1097/tp.0000000000002058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Aspergillus colonization after lung transplant is associated with an increased risk of chronic lung allograft dysfunction (CLAD). We hypothesized that gene expression during Aspergillus colonization could provide clues to CLAD pathogenesis. METHODS We examined transcriptional profiles in 3- or 6-month surveillance bronchoalveolar lavage fluid cell pellets from recipients with Aspergillus fumigatus colonization (n = 12) and without colonization (n = 10). Among the Aspergillus colonized, we also explored profiles in those who developed CLAD (n = 6) or remained CLAD-free (n = 6). Transcription profiles were assayed with the HG-U133 Plus 2.0 microarray (Affymetrix). Differential gene expression was based on an absolute fold difference of 2.0 or greater and unadjusted P value less than 0.05. We used NIH Database for Annotation, Visualization and Integrated Discovery for functional analyses, with false discovery rates less than 5% considered significant. RESULTS Aspergillus colonization was associated with differential expression of 489 probe sets, representing 404 unique genes. "Defense response" genes and genes in the "cytokine-cytokine receptor" Kyoto Encyclopedia of Genes and Genomes pathway were notably enriched in this list. Among Aspergillus colonized patients, CLAD development was associated with differential expression of 69 probe sets, representing 64 unique genes. This list was enriched for genes involved in "immune response" and "response to wounding", among others. Notably, both chitinase 3-like-1 and chitotriosidase were associated with progression to CLAD. CONCLUSIONS Aspergillus colonization is associated with gene expression profiles related to defense responses including cytokine signaling. Epithelial wounding, as well as the innate immune response to chitin that is present in the fungal cell wall, may be key in the link between Aspergillus colonization and CLAD.
Collapse
Affiliation(s)
- S Samuel Weigt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Xiaoyan Wang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Naman Patel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Ariss Derhovanessian
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Michael Y Shino
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - David M Sayah
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Joseph P Lynch
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - David J Ross
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Bernie M Kubak
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Abbas Ardehali
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Scott Palmer
- Department of Medicine, Duke University, Durham, NC
| | - Shahid Husain
- Department of Medicine, Division of Infectious Diseases, University of Toronto, Toronto, Ontario, Canada
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
56
|
Geube M, Anandamurthy B, Yared JP. Perioperative Management of the Lung Graft Following Lung Transplantation. Crit Care Clin 2018; 35:27-43. [PMID: 30447779 DOI: 10.1016/j.ccc.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Perioperative management of patients undergoing lung transplantation is one of the most complex in cardiothoracic surgery. Certain perioperative interventions, such as mechanical ventilation, fluid management and blood transfusions, use of extracorporeal mechanical support, and pain management, may have significant impact on the lung graft function and clinical outcome. This article provides a review of perioperative interventions that have been shown to impact the perioperative course after lung transplantation.
Collapse
Affiliation(s)
- Mariya Geube
- Department of Cardiothoracic Anesthesiology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland Clinic, 9500 Euclid Avenue, J4-331, Cleveland, OH 44195, USA.
| | - Balaram Anandamurthy
- Department of Cardiothoracic Anesthesiology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland Clinic, 9500 Euclid Avenue, J4-331, Cleveland, OH 44195, USA
| | - Jean-Pierre Yared
- Department of Cardiothoracic Anesthesiology, Cleveland Clinic, 9500 Euclid Avenue, J4-331, Cleveland, OH 44195, USA
| |
Collapse
|
57
|
Siddiqui AS, Kumar G, Majumdar T, Graviss EA, Nguyen DT, Goodarzi A, Kaleekal T. Association of methacholine challenge test with diagnosis of chronic lung allograft dysfunction in lung transplant patients. Clin Transplant 2018; 32:e13397. [PMID: 30192029 DOI: 10.1111/ctr.13397] [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: 12/08/2017] [Revised: 08/08/2018] [Accepted: 08/23/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) is a complication of lung transplantation. We sought to determine whether bronchial hyperresponsiveness detected by the methacholine challenge test (MCT) at 3 months after lung transplant (LT) predicts the development of CLAD. METHODS We performed a retrospective cohort study of 140 LT patients between 1/2008 and 6/2014 who underwent MCT at 3 months after LT. Pearson's chi-squared test and Kruskal-Wallis test were used to compare categorical and continuous variables, respectively. Cox proportional hazards modeling was used to evaluate the association between CLAD and MCT. RESULTS Methacholine challenge test+ was associated with the development of overall CLAD (adjusted hazards ratio [aHR]: 3.47; 95% confidence interval [95% CI]: 1.71, 7.03; P = 0.001) and CLAD within 3 years (aHR: 4.98; 95%CI: 1.84, 13.48; P = 0.002). Subgroup analysis showed that MCT (+) is associated with overall CLAD in single lung transplant (SLT) (aHR: 8.18; 95% CI: 2.22, 30.09; P = 0.002), double lung transplant (DLT) (aHR: 3.27; 95% CI: 1.22, 8.78; P = 0.02) and CLAD within 3 years in DLT patients (aHR: 6.76; 95% CI: 1.71, 26.74; P = 0.01). CONCLUSION Methacholine challenge test+ at 3 months after LT is associated with the development of overall CLAD. Positive MCT could predict the development of early CLAD within 3 years in DLT patients.
Collapse
Affiliation(s)
- Atif S Siddiqui
- Pulmonary and Critical Care, Houston Methodist Hospital, Houston, Texas
| | - Gagan Kumar
- Pulmonary and Critical Care, Northeast Georgia Medical Center, Gainesville, Georgia
| | | | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Duc T Nguyen
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Ahmad Goodarzi
- Pulmonary Transplant, Houston Methodist Hospital, Houston, Texas
| | - Thomas Kaleekal
- Pulmonary Transplant, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
58
|
Hoek RAS, Gaitanakis S, Hellemons ME. Insights from the European Respiratory Society 2018 Annual International Congress in the fields of thoracic surgery and lung transplantation. J Thorac Dis 2018; 10:S3005-S3009. [PMID: 30310690 PMCID: PMC6174134 DOI: 10.21037/jtd.2018.09.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Rogier A. S. Hoek
- Department of Pulmonary Medicine, division of lung transplantation, Erasmus Medical Center Rotterdam, The Netherlands
| | | | - Merel E. Hellemons
- Department of Pulmonary Medicine, division of lung transplantation, Erasmus Medical Center Rotterdam, The Netherlands
| |
Collapse
|
59
|
Luc JGY, Jackson K, Weinkauf JG, Freed DH, Nagendran J. Feasibility of Lung Transplantation From Donation After Circulatory Death Donors Following Portable Ex Vivo Lung Perfusion: A Pilot Study. Transplant Proc 2018; 49:1885-1892. [PMID: 28923643 DOI: 10.1016/j.transproceed.2017.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/03/2017] [Accepted: 04/27/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Donation after circulatory death (DCD) has the potential to significantly alleviate the shortage of transplantable lungs. We report our initial experience with the use of portable ex vivo lung perfusion (EVLP) with the Organ Care System Lung device for evaluation of DCD lungs. METHODS We performed a retrospective review of the DCD lung transplantation (LTx) experience at a single institution through the use of a prospective database. RESULTS From 2011 to 2015, 208 LTx were performed at the University of Alberta, of which 11 were DCD LTx with 7 (64%) that underwent portable EVLP. DCD lungs preserved with portable EVLP had a significantly shorter cold ischemic time (161 ± 44 vs 234 ± 60 minutes, P = .045), lower grade of primary graft dysfunction at 72 hours after LTx (0.4 ± 0.5 vs 2.1 ± 0.7, P = .003), similar mechanical ventilation time (55 ± 44 vs 103 ± 97 hours, P = .281), and hospital length of stay (29 ± 11 vs 33 ± 10 days, P = .610). All patients were alive at 1-year follow-up after LTx with improved functional outcomes and acceptable quality of life compared with before LTx, although there were no intergroup differences. CONCLUSIONS In our pilot cohort, portable EVLP was a feasible modality to increase confidence in the use of DCD lungs with validated objective evidence of lung function during EVLP that translates to acceptable clinical outcomes and quality of life after LTx. Further studies are needed to validate these initial findings in a larger cohort.
Collapse
Affiliation(s)
- J G Y Luc
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, Edmonton, Canada
| | - K Jackson
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - J G Weinkauf
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - D H Freed
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, Edmonton, Canada; Alberta Transplant Institute, Edmonton, Canada; Canadian National Transplant Research Program, Edmonton, Canada
| | - J Nagendran
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, Edmonton, Canada; Alberta Transplant Institute, Edmonton, Canada; Canadian National Transplant Research Program, Edmonton, Canada.
| |
Collapse
|
60
|
Normothermic ex-vivo preservation with the portable Organ Care System Lung device for bilateral lung transplantation (INSPIRE): a randomised, open-label, non-inferiority, phase 3 study. THE LANCET RESPIRATORY MEDICINE 2018; 6:357-367. [DOI: 10.1016/s2213-2600(18)30136-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/28/2022]
|
61
|
Verleden SE, Martens A, Ordies S, Neyrinck AP, Van Raemdonck DE, Verleden GM, Vanaudenaerde BM, Vos R. Immediate post-operative broncho-alveolar lavage IL-6 and IL-8 are associated with early outcomes after lung transplantation. Clin Transplant 2018; 32:e13219. [PMID: 29405435 DOI: 10.1111/ctr.13219] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2018] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Previous studies demonstrated that increased cytokine and chemokine levels, either shortly before or after lung transplantation, were associated with post-transplant outcome. However, small patient cohorts were mostly used, focusing on 1 molecule and 1 outcome. In a large single-center cohort, we investigated the predictive value of immediate post-operative broncho-alveolar lavage (BAL) expression of IL-6 and IL-8 on multiple key outcomes, including PGD, CLAD, graft survival, as well as several secondary outcomes. MATERIAL AND METHODS All patients undergoing a first lung transplant in whom routine bronchoscopy with BAL was performed during the first 48 hours post-transplantation were included. IL-6 and IL-8 protein levels were measured in BAL via ELISA. RESULTS A total of 336 patients were included. High IL-6 levels measured within 24 hours of transplantation were associated with longer time on ICU and time to hospital discharge; and increased prevalence of PGD grade 3. Increased IL-8 levels, measured within 24 hours, were associated with PGD3, more ECMO use, higher donor paO2 , younger donor age, but not with other short-or long-term outcome. IL-6 and IL-8 measured between 24 and 48 hours of transplantation were not associated with any outcome parameters. CONCLUSION Recipient BAL IL-6 and IL-8 within 24 hours post-transplant were associated with an increased incidence of PGD3.
Collapse
Affiliation(s)
- Stijn E Verleden
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - An Martens
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Sofie Ordies
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of cardiovascular sciences, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung transplant unit, Department of chronic diseases, metabolism and ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
62
|
Cantu E, Diamond JM, Suzuki Y, Lasky J, Schaufler C, Lim B, Shah R, Porteous M, Lederer DJ, Kawut SM, Palmer SM, Snyder LD, Hartwig MG, Lama VN, Bhorade S, Bermudez C, Crespo M, McDyer J, Wille K, Orens J, Shah PD, Weinacker A, Weill D, Wilkes D, Roe D, Hage C, Ware LB, Bellamy SL, Christie JD. Quantitative Evidence for Revising the Definition of Primary Graft Dysfunction after Lung Transplant. Am J Respir Crit Care Med 2018; 197:235-243. [PMID: 28872353 PMCID: PMC5768905 DOI: 10.1164/rccm.201706-1140oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/01/2017] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Primary graft dysfunction (PGD) is a form of acute lung injury that occurs after lung transplantation. The definition of PGD was standardized in 2005. Since that time, clinical practice has evolved, and this definition is increasingly used as a primary endpoint for clinical trials; therefore, validation is warranted. OBJECTIVES We sought to determine whether refinements to the 2005 consensus definition could further improve construct validity. METHODS Data from the Lung Transplant Outcomes Group multicenter cohort were used to compare variations on the PGD definition, including alternate oxygenation thresholds, inclusion of additional severity groups, and effects of procedure type and mechanical ventilation. Convergent and divergent validity were compared for mortality prediction and concurrent lung injury biomarker discrimination. MEASUREMENTS AND MAIN RESULTS A total of 1,179 subjects from 10 centers were enrolled from 2007 to 2012. Median length of follow-up was 4 years (interquartile range = 2.4-5.9). No mortality differences were noted between no PGD (grade 0) and mild PGD (grade 1). Significantly better mortality discrimination was evident for all definitions using later time points (48, 72, or 48-72 hours; P < 0.001). Biomarker divergent discrimination was superior when collapsing grades 0 and 1. Additional severity grades, use of mechanical ventilation, and transplant procedure type had minimal or no effect on mortality or biomarker discrimination. CONCLUSIONS The PGD consensus definition can be simplified by combining lower PGD grades. Construct validity of grading was present regardless of transplant procedure type or use of mechanical ventilation. Additional severity categories had minimal impact on mortality or biomarker discrimination.
Collapse
Affiliation(s)
| | - Joshua M. Diamond
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | | | - Brian Lim
- Division of Cardiovascular Surgery and
| | - Rupal Shah
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Mary Porteous
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - David J. Lederer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Steven M. Kawut
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Clinical Epidemiology and Biostatistics and
- Penn Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Scott M. Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | | | - Matthew G. Hartwig
- Division of Cardiothoracic Surgery, Duke University, Durham, North Carolina
| | - Vibha N. Lama
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sangeeta Bhorade
- Division of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | | | - Maria Crespo
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - John McDyer
- Division of Pulmonary, Allergy, and Critical Care, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Keith Wille
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jonathan Orens
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Pali D. Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Ann Weinacker
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - David Weill
- Institute for Advanced Organ Disease and Transplantation, University of South Florida, Tampa, Florida
| | - David Wilkes
- Division of Pulmonary, Allergy, Critical Care, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - David Roe
- Division of Pulmonary, Allergy, Critical Care, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Chadi Hage
- Division of Pulmonary, Allergy, Critical Care, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lorraine B. Ware
- Department of Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; and
| | - Scarlett L. Bellamy
- Dornsife School of Public Health, Drexel University, Philadelphia, Pennsylvania
| | - Jason D. Christie
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Clinical Epidemiology and Biostatistics and
| |
Collapse
|
63
|
Hamacher J, Hadizamani Y, Borgmann M, Mohaupt M, Männel DN, Moehrlen U, Lucas R, Stammberger U. Cytokine-Ion Channel Interactions in Pulmonary Inflammation. Front Immunol 2018; 8:1644. [PMID: 29354115 PMCID: PMC5758508 DOI: 10.3389/fimmu.2017.01644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
The lungs conceptually represent a sponge that is interposed in series in the bodies’ systemic circulation to take up oxygen and eliminate carbon dioxide. As such, it matches the huge surface areas of the alveolar epithelium to the pulmonary blood capillaries. The lung’s constant exposure to the exterior necessitates a competent immune system, as evidenced by the association of clinical immunodeficiencies with pulmonary infections. From the in utero to the postnatal and adult situation, there is an inherent vital need to manage alveolar fluid reabsorption, be it postnatally, or in case of hydrostatic or permeability edema. Whereas a wealth of literature exists on the physiological basis of fluid and solute reabsorption by ion channels and water pores, only sparse knowledge is available so far on pathological situations, such as in microbial infection, acute lung injury or acute respiratory distress syndrome, and in the pulmonary reimplantation response in transplanted lungs. The aim of this review is to discuss alveolar liquid clearance in a selection of lung injury models, thereby especially focusing on cytokines and mediators that modulate ion channels. Inflammation is characterized by complex and probably time-dependent co-signaling, interactions between the involved cell types, as well as by cell demise and barrier dysfunction, which may not uniquely determine a clinical picture. This review, therefore, aims to give integrative thoughts and wants to foster the unraveling of unmet needs in future research.
Collapse
Affiliation(s)
- Jürg Hamacher
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Internal Medicine V - Pneumology, Allergology, Respiratory and Environmental Medicine, Faculty of Medicine, Saarland University, Saarbrücken, Germany.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Yalda Hadizamani
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Michèle Borgmann
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Markus Mohaupt
- Internal Medicine, Sonnenhofspital Bern, Bern, Switzerland
| | | | - Ueli Moehrlen
- Paediatric Visceral Surgery, Universitäts-Kinderspital Zürich, Zürich, Switzerland
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Vascular Biology Center, Medical College of Georgia, Augusta, GA, United States
| | - Uz Stammberger
- Lungen- und Atmungsstiftung Bern, Bern, Switzerland.,Novartis Institutes for Biomedical Research, Translational Clinical Oncology, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
64
|
Pentraxin 3 deficiency enhances features of chronic rejection in a mouse orthotopic lung transplantation model. Oncotarget 2018; 9:8489-8501. [PMID: 29492210 PMCID: PMC5823599 DOI: 10.18632/oncotarget.23902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/12/2017] [Indexed: 12/24/2022] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is a serious complication after lung transplantation and thought to represent chronic rejection. Increased expression of Pentraxin 3 (PTX3), an acute phase protein, was associated with worse outcome in lung transplant patients. To determine the role of recipient PTX3 in development of chronic rejection, we used a minor alloantigen-mismatched murine orthotopic single lung transplant model. Male C57BL/10 mice were used as donors. Male PTX3 knockout (KO) mice and their wild type (WT) littermates on 129/SvEv/C57BL6/J background were used as recipients. In KO recipients, 7/13 grafted lungs were consolidated without volume recovery on CT scan, while only 2/9 WT mice showed similar graft consolidation. For grafts where lung volume could be reliably analyzed by CT scan, the lung volume recovery was significantly reduced in KO mice compared to WT. Interstitial inflammation, parenchymal fibrosis and bronchiolitis obliterans scores were significantly higher in KO mice. Presence of myofibroblasts and lymphoid aggregation was significantly enhanced in the grafts of PTX3 KO recipients. Recipient PTX3 deficiency enhanced chronic rejection-like lesions by promoting a fibrotic process in the airways and lung parenchyma. The underlying mechanisms and potential protective role of exogenous PTX3 as a therapy should be further explored.
Collapse
|
65
|
Abstract
Primary graft dysfunction is a form of acute injury after lung transplantation that is associated with significant short- and long-term morbidity and mortality. Multiple mechanisms contribute to the pathogenesis of primary graft dysfunction, including ischemia reperfusion injury, epithelial cell death, endothelial cell dysfunction, innate immune activation, oxidative stress, and release of inflammatory cytokines and chemokines. This article reviews the epidemiology, pathogenesis, risk factors, prevention, and treatment of primary graft dysfunction.
Collapse
Affiliation(s)
- Mary K Porteous
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA 19104, USA.
| | - James C Lee
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
66
|
Diamond JM, Arcasoy S, Kennedy CC, Eberlein M, Singer JP, Patterson GM, Edelman JD, Dhillon G, Pena T, Kawut SM, Lee JC, Girgis R, Dark J, Thabut G. Report of the International Society for Heart and Lung Transplantation Working Group on Primary Lung Graft Dysfunction, part II: Epidemiology, risk factors, and outcomes—A 2016 Consensus Group statement of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2017; 36:1104-1113. [DOI: 10.1016/j.healun.2017.07.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 11/28/2022] Open
|
67
|
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
|
68
|
Report of the ISHLT Working Group on Primary Lung Graft Dysfunction, part I: Definition and grading-A 2016 Consensus Group statement of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2017; 36:1097-1103. [PMID: 28942784 DOI: 10.1016/j.healun.2017.07.021] [Citation(s) in RCA: 447] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/27/2022] Open
|
69
|
Gelman AE, Fisher AJ, Huang HJ, Baz MA, Shaver CM, Egan TM, Mulligan MS. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction Part III: Mechanisms: A 2016 Consensus Group Statement of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2017; 36:1114-1120. [PMID: 28818404 DOI: 10.1016/j.healun.2017.07.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/16/2017] [Indexed: 01/17/2023] Open
Affiliation(s)
- Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.
| | - Andrew J Fisher
- Institute of Transplantation, Freeman Hospital and Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Howard J Huang
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas, USA
| | - Maher A Baz
- Departments of Medicine and Surgery, University of Kentucky, Lexington, Kentucky, USA
| | - Ciara M Shaver
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Thomas M Egan
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Micheal S Mulligan
- Department of Surgery, Division of Cardiothoracic Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
70
|
Glanville AR. Physiology of chronic lung allograft dysfunction: back to the future? Eur Respir J 2017; 49:49/4/1700187. [DOI: 10.1183/13993003.00187-2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 11/05/2022]
|
71
|
Zeriouh M, Sabashnikov A, Mohite PN, Zych B, Patil NP, García-Sáez D, Koch A, Weymann A, Soresi S, Wippermann J, Wahlers T, De Robertis F, Popov AF, Simon AR. Zonal organ allocation system and its impact on long-term outcomes after lung transplantation: a propensity score matched analysis†. Eur J Cardiothorac Surg 2016; 51:119-126. [PMID: 27694251 DOI: 10.1093/ejcts/ezw284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/05/2016] [Accepted: 05/26/2016] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Zonal organ allocation system comprises organ procurement by teams within a specific geographical area of each retrieval team. Therefore, in a substantial number of cases organs are retrieved by 'foreign' teams and are sent for transplantation to the implanting centre. The aim of this study was to assess the impact of zonal organ allocation system on early- and long-term outcomes after lung transplantation (LTx). METHODS Included were 331 consecutive patients who underwent LTx performed at Harefield Hospital between January 2007 and January 2015. Recipients were divided into two groups depending on the organ retrieval team: 204 (61.6%) patients were transplanted using lungs procured by our institutional team (institutional group), whereas 127 (38.4%) organs were retrieved by other teams (external group) from experienced transplant centres in the UK. To exclude selection bias and other confounders, a 1:1 propensity score-based matching procedure was performed resulting in a total number of 238 donors and recipients who were well matched for baseline characteristics. The primary end-points were overall survival after LTx and freedom from bronchiolitis obliterans syndrome (BOS). Secondary end-points were perioperative clinical characteristics as well as adverse events that occurred over the follow-up. RESULT After propensity score matching all donor characteristics and all baseline recipient characteristics were statistically similar between the two groups. In terms of early postoperative results, both groups were statistically comparable. However, there was a trend towards higher incidence of primary graft dysfunction in the external group (P = 0.054). Regarding long-term results with up to 7 years of follow-up, the overall survival also appeared to be poorer in the external group; however, this difference did not reach statistical significance. The freedom from BOS over the long-term follow was significantly poorer in the external group (P = 0.040). CONCLUSION Despite excellent early outcomes the zonal allocation system might be associated with significantly poorer long-term outcomes in terms of freedom from BOS after bilateral LTx. Further research is needed to find the underlying factors leading to these results.
Collapse
Affiliation(s)
- Mohamed Zeriouh
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK.,Department of Cardiothoracic Surgery, University Hospital of Cologne, Cologne, Germany
| | - Anton Sabashnikov
- Department of Cardiothoracic Surgery, University Hospital of Cologne, Cologne, Germany
| | - Prashant N Mohite
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Bartlomiej Zych
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Nikhil P Patil
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Diana García-Sáez
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Achim Koch
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Alexander Weymann
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Simona Soresi
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Jens Wippermann
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Thorsten Wahlers
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Fabio De Robertis
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Aron-Frederik Popov
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - André R Simon
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| |
Collapse
|
72
|
Bharat A, Chiu S, Zheng Z, Sun H, Yeldandi A, DeCamp MM, Perlman H, Budinger GRS, Mohanakumar T. Lung-Restricted Antibodies Mediate Primary Graft Dysfunction and Prevent Allotolerance after Murine Lung Transplantation. Am J Respir Cell Mol Biol 2016; 55:532-541. [PMID: 27144500 DOI: 10.1165/rcmb.2016-0077oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Over one-third of lung recipients have preexisting antibodies against lung-restricted antigens: collagen (Col) type V and K-α1 tubulin (KAT). Although clinical studies have shown association of these antibodies with primary graft dysfunction (PGD), their biological significance remains unclear. We tested whether preexisting lung-restricted antibodies can mediate PGD and prevent allotolerance. A murine syngeneic (C57BL/6) or allogeneic (C57BL/6 to BALB/c) left lung transplantation model was used. Rabbit polyclonal antibodies were produced against KAT and Col-V and injected pretransplantation. T cell frequency was analyzed using enzyme-linked immunospot, whereas alloantibodies were determined using flow cytometry. Wet:dry ratio, arterial oxygenation, and histology were used to determine PGD. Preexisting Col-V or KAT, but not isotype control, antibodies lead to dose-dependent development of PGD after syngeneic lung transplantation, as evidenced by poor oxygenation and increased wet:dry ratio. Histology confirmed alveolar and capillary edema. The native right lung remained unaffected. Epitope spreading was observed where KAT antibody treatment led to the development of IL-17-producing CD4+ T cells and humoral response against Col-V, or vice versa. In contrast, isotype control antibody failed to induce Col-V- or KAT-specific cellular or humoral immunity. In addition, none of the mice developed immunity against a non-lung antigen, collagen type II. Preexisting lung-restricted antibodies, but not isotype control, prevented development of allotolerance using the MHC-related 1 and cytotoxic T-lymphocyte-associated protein 4-Ig regimen. Lung-restricted antibodies can induce both early and delayed lung graft dysfunction. These antibodies can also cause spreading of lung-restricted immunity and promote alloimmunity. Antibody-directed therapy to treat preexisting lung-restricted antibodies might reduce PGD after lung transplantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Harris Perlman
- 3 Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | - G R Scott Budinger
- 3 Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | | |
Collapse
|
73
|
Olland A, Reeb J, Leclerq A, Renaud-Picard B, Falcoz PE, Kessler R, Schini-Kerth V, Kessler L, Toti F, Massard G. Microparticles: A new insight into lung primary graft dysfunction? Hum Immunol 2016; 77:1101-1107. [PMID: 27381358 DOI: 10.1016/j.humimm.2016.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 04/17/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022]
Abstract
Lung transplantation is the only life-saving treatment for end stage respiratory disease. The immediate outcome is still hampered by primary graft dysfunction. The latter is a form of acute lung injury occurring within the 30min following the unclamping of the pulmonary artery that prompts ischemia reperfusion injury. Severe forms may need prolonged mechanical ventilation and extra-corporeal membrane oxygenation. Overall, primary graft dysfunction accounts for at least one third of the deaths during the first post-operative month. Despite increasing experience and knowledge on the underlying cellular events, there is still a lack of an early marker of ischemia reperfusion graft injuries. Microparticles are plasma membrane vesicles that are released from damaged or stressed cells in biological fluids and remodeling tissues, among which the lung parenchyma during acute or chronic injury. We recently evidenced alveolar microparticles as surrogate markers of strong ischemia injury in ex-vivo reperfusion experimental models. We propose herein new insights on how microparticles may be helpful to evaluate the extent of lung ischemia reperfusion injuries and predict the occurrence of primary graft dysfunction.
Collapse
Affiliation(s)
- Anne Olland
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France.
| | - Jérémie Reeb
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Alexandre Leclerq
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Benjamin Renaud-Picard
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Pierre-Emmanuel Falcoz
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Romain Kessler
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Valérie Schini-Kerth
- UMR CNRS 7213, Biophotonique and Pharmacology Laboratory, Pharmacology School, University of Strasbourg, Strasbourg, France
| | - Laurence Kessler
- Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| | - Florence Toti
- UMR CNRS 7213, Biophotonique and Pharmacology Laboratory, Pharmacology School, University of Strasbourg, Strasbourg, France
| | - Gilbert Massard
- Labex Transplantex, Translational Medecine Federation, University of Strasbourg, Strasbourg, France; Lung Transplantation Group, University Hospital Strasbourg, Strasbourg France; EA 7293 SVTT 'Stress Vasculaire et Tissulaire en Transplantation', Translational Medecine Federation, University of Strasbourg, Strasbourg, France
| |
Collapse
|
74
|
Daimiel Naranjo I, Alonso Charterina S. What can happen after lung transplantation and the importance of the time since transplantation: Radiological review of post-transplantation complications. RADIOLOGIA 2016. [DOI: 10.1016/j.rxeng.2016.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
75
|
Hashimoto K, Besla R, Zamel R, Juvet S, Kim H, Azad S, Waddell TK, Cypel M, Liu M, Keshavjee S. Circulating Cell Death Biomarkers May Predict Survival in Human Lung Transplantation. Am J Respir Crit Care Med 2016; 194:97-105. [DOI: 10.1164/rccm.201510-2115oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
76
|
Daimiel Naranjo I, Alonso Charterina S. What can happen after lung transplantation and the importance of the time since transplantation: radiological review of post-transplantation complications. RADIOLOGIA 2016; 58:257-67. [PMID: 27017046 DOI: 10.1016/j.rx.2016.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/10/2016] [Accepted: 02/13/2016] [Indexed: 11/18/2022]
Abstract
Lung transplantation is the best treatment option in the final stages of diseases such as cystic fibrosis, pulmonary hypertension, chronic obstructive pulmonary disease, or idiopathic pulmonary fibrosis. Better surgical techniques and advances in immunosuppressor treatments have increased survival in lung transplant recipients, making longer follow-up necessary because complications can occur at any time after transplantation. For practical purposes, complications can be classified as early (those that normally occur within two months after transplantation), late (those that normally occur more than two months after transplantation), or time-independent (those that can occur at any time after transplantation). Many complications have nonspecific clinical and radiological manifestations, so the time factor is key to narrow the differential diagnosis. Imaging can guide interventional procedures and can detect complications early. This article aims to describe and illustrate the complications that can occur after lung transplantation from the clinical and radiological viewpoints so that they can be detected as early as possible.
Collapse
Affiliation(s)
- I Daimiel Naranjo
- Servicio de Radiodiagnóstico, Hospital Universitario 12 de Octubre, Madrid, España.
| | - S Alonso Charterina
- Servicio de Radiodiagnóstico, Hospital Universitario 12 de Octubre, Madrid, España
| |
Collapse
|
77
|
Smibert O, Snell GI, Bills H, Westall GP, Morrissey CO. Mycobacterium abscessus Complex - a Particular Challenge in the Setting of Lung Transplantation. Expert Rev Anti Infect Ther 2016; 14:325-33. [PMID: 26732819 DOI: 10.1586/14787210.2016.1138856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mycobacterium abscessus complex is an emerging pathogen in lung transplant candidates and recipients. M. abscessus complex is widespread in the environment and can cause pulmonary, skin and soft tissue, and disseminated infection, particularly in lung transplant recipients. It is innately resistant to many antibiotics making it difficult to treat. Herein we describe the epidemiology, clinical manifestations, diagnosis and treatment of M. abscessus with an emphasis on lung transplant candidates and recipients. We also outline the areas where data are lacking and the areas where further research is urgently needed.
Collapse
Affiliation(s)
- O Smibert
- a Department of Infectious Diseases , Alfred Health and Monash University , Melbourne , Australia
| | - G I Snell
- b Department of Allergy, Immunology and Respiratory Medicine , Alfred Health and Monash University , Melbourne , Australia
| | - H Bills
- c Faculty of Medicine , Nursing and Health Sciences, Monash University , Clayton , Australia
| | - G P Westall
- b Department of Allergy, Immunology and Respiratory Medicine , Alfred Health and Monash University , Melbourne , Australia
| | - C O Morrissey
- a Department of Infectious Diseases , Alfred Health and Monash University , Melbourne , Australia
| |
Collapse
|
78
|
DerHovanessian A, Weigt SS, Palchevskiy V, Shino MY, Sayah DM, Gregson AL, Noble PW, Palmer SM, Fishbein MC, Kubak BM, Ardehali A, Ross DJ, Saggar R, Lynch JP, Elashoff RM, Belperio JA. The Role of TGF-β in the Association Between Primary Graft Dysfunction and Bronchiolitis Obliterans Syndrome. Am J Transplant 2016; 16:640-9. [PMID: 26461171 PMCID: PMC4946573 DOI: 10.1111/ajt.13475] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 06/09/2015] [Accepted: 07/03/2015] [Indexed: 01/25/2023]
Abstract
Primary graft dysfunction (PGD) is a possible risk factor for bronchiolitis obliterans syndrome (BOS) following lung transplantation; however, the mechanism for any such association is poorly understood. Based on the association of TGF-β with acute and chronic inflammatory disorders, we hypothesized that it might play a role in the continuum between PGD and BOS. Thus, the association between PGD and BOS was assessed in a single-center cohort of lung transplant recipients. Bronchoalveolar lavage fluid concentrations of TGF-β and procollagen collected within 24 h of transplantation were compared across the spectrum of PGD, and incorporated into Cox models of BOS. Immunohistochemistry localized expression of TGF-β and its receptor in early lung biopsies posttransplant. We found an association between PGD and BOS in both bilateral and single lung recipients with a hazard ratio of 3.07 (95% CI 1.76-5.38) for the most severe form of PGD. TGF-β and procollagen concentrations were elevated during PGD (p < 0.01), and associated with increased rates of BOS. Expression of TGF-β and its receptor localized to allograft infiltrating mononuclear and stromal cells, and the airway epithelium. These findings validate the association between PGD and the subsequent development of BOS, and suggest that this association may be mediated by receptor/TGF-β biology.
Collapse
Affiliation(s)
- Ariss DerHovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - S. Samuel Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Vyacheslav Palchevskiy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Michael Y. Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - David M. Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Aric L. Gregson
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California
| | - Paul W. Noble
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles California
| | - Scott M. Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University, Durham, North Carolina
| | - Michael C. Fishbein
- Depatment of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Bernard M. Kubak
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California
| | - Abbas Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, Los Angeles, California
| | - David J. Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Joseph P. Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Robert M. Elashoff
- Department of Biomathematics, University of California, Los Angeles, California
| | - John A. Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| |
Collapse
|
79
|
Lung Transplantation. PATHOLOGY OF TRANSPLANTATION 2016. [PMCID: PMC7153460 DOI: 10.1007/978-3-319-29683-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The therapeutic options for patients with advanced pulmonary parenchymal or vascular disorders are currently limited. Lung transplantation remains one of the few viable interventions, but on account of the insufficient donor pool only a minority of these patients actually undergo the procedure each year. Following transplantation there are a number of early and late allograft complications such as primary graft dysfunction, allograft rejection, infection, post-transplant lymphoproliferative disorder and late injury that is now classified as chronic lung allograft dysfunction. The pathologist plays an essential role in the diagnosis and classification of these myriad complications. Although the transplant procedures are performed in selected centers patients typically return to their local centers. When complications arise it is often the responsibility of the local pathologist to evaluate specimens. Therefore familiarity with the pathology of lung transplantation is important.
Collapse
|
80
|
Porteous MK, Diamond JM, Christie JD. Primary graft dysfunction: lessons learned about the first 72 h after lung transplantation. Curr Opin Organ Transplant 2015; 20:506-14. [PMID: 26262465 PMCID: PMC4624097 DOI: 10.1097/mot.0000000000000232] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW In 2005, the International Society for Heart and Lung Transplantation published a standardized definition of primary graft dysfunction (PGD), facilitating new knowledge on this form of acute lung injury that occurs within 72 h of lung transplantation. PGD continues to be associated with significant morbidity and mortality. This article will summarize the current literature on the epidemiology of PGD, pathogenesis, risk factors, and preventive and treatment strategies. RECENT FINDINGS Since 2011, several manuscripts have been published that provide insight into the clinical risk factors and pathogenesis of PGD. In addition, several transplant centers have explored preventive and treatment strategies for PGD, including the use of extracorporeal strategies. More recently, results from several trials assessing the role of extracorporeal lung perfusion may allow for much-needed expansion of the donor pool, without raising PGD rates. SUMMARY This article will highlight the current state of the science regarding PGD, focusing on recent advances, and set a framework for future preventive and treatment strategies.
Collapse
Affiliation(s)
- Mary K Porteous
- aDepartment of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA bCenter for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
81
|
Role of innate immunity in primary graft dysfunction after lung transplantation. Curr Opin Organ Transplant 2015; 18:518-23. [PMID: 23995372 DOI: 10.1097/mot.0b013e3283651994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD), a form of acute lung injury after lung transplantation, has a significant impact on clinical outcomes after lung transplantation. This potentially reversible graft impairment occurs after ischemia-reperfusion injury. This review describes the expanding body of literature evaluating the central role of innate immune activation, nonadaptive responses and dysregulation in the development of PGD after lung transplant. RECENT FINDINGS The innate immune system, highlighted by Toll-like receptor pathways and neutrophil migration and influx, plays an important role in the initiation and propagation of ischemia-reperfusion injury. Recent plasma biomarker and gene association studies have identified several genes and proteins composing innate immune pathways to be associated with PGDs. Long pentraxin-3 and Toll-like receptors, as well as inflammasomes and Toll-interacting protein, are associated with the development of PGD after lung transplantation. SUMMARY Innate immune pathways are involved in the development of PGD and may provide attractive targets for therapies. It may be possible to prevent or treat PGD, as well as to allow pre-transplant PGD risk stratification. To improve understanding of the mechanisms behind clinical risk factors for PGD will require further in-depth correlation of donor-specific and recipient-related triggers of nonadaptive immune responses.
Collapse
|
82
|
Agashe VV, Burlingham WJ. Autoimmune Reactivity in Graft Injury: Player or Bystander? CURRENT TRANSPLANTATION REPORTS 2015; 2:211-221. [PMID: 29057202 DOI: 10.1007/s40472-015-0068-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Organ transplantation is the only viable treatment for several end-stage organ failures. However chronic rejection prevents long-term graft survival. Traditionally this rejection was attributed to the development of alloimmunity in transplant patients. However recent evidence suggests that autoimmunity plays a larger role in chronic rejection of certain organ transplants, than alloimmunity. In this review we will focus on the history of autoimmunity in solid-organ transplantation and at look the Collagen Type V, K-α-tubulin, Vimentin, Cardiac myosin and Heat Shock Proteins as classical examples of auto-antigens in organ transplantation. We will also look at some of the recent reports looking at the mechanisms of autoimmunity and try to provide answers to some of the age-old questions in autoimmunity.
Collapse
Affiliation(s)
- Vrushali V Agashe
- Comparative Biomedical Sciences Graduate Program.,Department of Surgery-Transplant division, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI 53795, USA
| | - William J Burlingham
- Department of Surgery-Transplant division, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI 53795, USA
| |
Collapse
|
83
|
Sayah DM, Mallavia B, Liu F, Ortiz-Muñoz G, Caudrillier A, DerHovanessian A, Ross DJ, Lynch JP, Saggar R, Ardehali A, Ware LB, Christie JD, Belperio JA, Looney MR. Neutrophil extracellular traps are pathogenic in primary graft dysfunction after lung transplantation. Am J Respir Crit Care Med 2015; 191:455-63. [PMID: 25485813 DOI: 10.1164/rccm.201406-1086oc] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Primary graft dysfunction (PGD) causes early mortality after lung transplantation and may contribute to late graft failure. No effective treatments exist. The pathogenesis of PGD is unclear, although both neutrophils and activated platelets have been implicated. We hypothesized that neutrophil extracellular traps (NETs) contribute to lung injury in PGD in a platelet-dependent manner. OBJECTIVES To study NETs in experimental models of PGD and in lung transplant patients. METHODS Two experimental murine PGD models were studied: hilar clamp and orthotopic lung transplantation after prolonged cold ischemia (OLT-PCI). NETs were assessed by immunofluorescence microscopy and ELISA. Platelet activation was inhibited with aspirin, and NETs were disrupted with DNaseI. NETs were also measured in bronchoalveolar lavage fluid and plasma from lung transplant patients with and without PGD. MEASUREMENTS AND MAIN RESULTS NETs were increased after either hilar clamp or OLT-PCI compared with surgical control subjects. Activation and intrapulmonary accumulation of platelets were increased in OLT-PCI, and platelet inhibition reduced NETs and lung injury, and improved oxygenation. Disruption of NETs by intrabronchial administration of DNaseI also reduced lung injury and improved oxygenation. In bronchoalveolar lavage fluid from human lung transplant recipients, NETs were more abundant in patients with PGD. CONCLUSIONS NETs accumulate in the lung in both experimental and clinical PGD. In experimental PGD, NET formation is platelet-dependent, and disruption of NETs with DNaseI reduces lung injury. These data are the first description of a pathogenic role for NETs in solid organ transplantation and suggest that NETs are a promising therapeutic target in PGD.
Collapse
Affiliation(s)
- David M Sayah
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Bhinder S, Chen H, Sato M, Copes R, Evans GJ, Chow CW, Singer LG. Air pollution and the development of posttransplant chronic lung allograft dysfunction. Am J Transplant 2014; 14:2749-57. [PMID: 25358842 DOI: 10.1111/ajt.12909] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 06/25/2014] [Accepted: 06/29/2014] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is the leading cause of mortality following lung transplantation. We conducted a retrospective cohort study including 397 bilateral lung recipients transplanted in from 1996 to 2009 to determine the association between ambient air pollution, CLAD and mortality. Pollution exposure was assessed using satellite-based estimates of nitrogen dioxide, distance to major roadway and total length of roadways around a patient's home. Cumulative exposures to ozone and particulate matter were estimated from concentrations measured at fixed-site stations near patients' homes using inverse distance weighted interpolation. Cox proportional hazards models were used to estimate the associations of CLAD with air pollution exposure, adjusting for various individual and neighborhood characteristics. During the follow-up, 185 patients developed CLAD (47%) and 101 patients died (25%). Fifty-four deaths (53%) were due to CLAD. We observed an association between CLAD development and road density within 200 m of a patient's home (HR 1.30 [95% CI 1.07-1.58]). Although based on a subgroup of 14 patients, living within 100 m of a highway was associated with a high risk for developing CLAD (HR 4.91 [95% CI 2.22, 10.87]). These data suggest that exposure to traffic-related air pollution is associated with development of CLAD among lung transplant recipients.
Collapse
Affiliation(s)
- S Bhinder
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
85
|
La Francesca S, Ting AE, Sakamoto J, Rhudy J, Bonenfant NR, Borg ZD, Cruz FF, Goodwin M, Lehman NA, Taggart JM, Deans R, Weiss DJ. Multipotent adult progenitor cells decrease cold ischemic injury in ex vivo perfused human lungs: an initial pilot and feasibility study. Transplant Res 2014; 3:19. [PMID: 25671090 PMCID: PMC4323223 DOI: 10.1186/2047-1440-3-19] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022] Open
Abstract
Background Primary graft dysfunction (PGD) is a significant cause of early morbidity and mortality following lung transplantation. Improved organ preservation techniques will decrease ischemia-reperfusion injury (IRI) contributing to PGD. Adult bone marrow-derived adherent stem cells, including mesenchymal stromal (stem) cells (MSCs) and multipotent adult progenitor cells (MAPCs), have potent anti-inflammatory actions, and we thus postulated that intratracheal MAPC administration during donor lung processing would decrease IRI. The goal of the study was therefore to determine if intratracheal MAPC instillation would decrease lung injury and inflammation in an ex vivo human lung explant model of prolonged cold storage and subsequent reperfusion. Methods Four donor lungs not utilized for transplant underwent 8 h of cold storage (4°C). Following rewarming for approximately 30 min, non-HLA-matched allogeneic MAPCs (1 × 107 MAPCs/lung) were bronchoscopically instilled into the left lower lobe (LLL) and vehicle comparably instilled into the right lower lobe (RLL). The lungs were then perfused and mechanically ventilated for 4 h and subsequently assessed for histologic injury and for inflammatory markers in bronchoalveolar lavage fluid (BALF) and lung tissue. Results All LLLs consistently demonstrated a significant decrease in histologic and BALF inflammation compared to vehicle-treated RLLs. Conclusions These initial pilot studies suggest that use of non-HLA-matched allogeneic MAPCs during donor lung processing can decrease markers of cold ischemia-induced lung injury. Electronic supplementary material The online version of this article (doi:10.1186/2047-1440-3-19) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Saverio La Francesca
- Cardiac Surgery and Cardiopulmonary Transplantation, DeBakey Heart and Vascular Center, The Houston Methodist, Houston, TX USA ; Harvard Apparatus Regenerative Technology, Inc, Holliston, MA USA
| | | | - Jason Sakamoto
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX USA
| | - Jessica Rhudy
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX USA
| | - Nicholas R Bonenfant
- Department of Medicine, University of Vermont College of Medicine, 226 Health Science Research Facility, Burlington, VT USA
| | - Zachary D Borg
- Department of Medicine, University of Vermont College of Medicine, 226 Health Science Research Facility, Burlington, VT USA
| | - Fernanda F Cruz
- Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Meagan Goodwin
- Department of Medicine, University of Vermont College of Medicine, 226 Health Science Research Facility, Burlington, VT USA
| | | | | | | | - Daniel J Weiss
- Department of Medicine, University of Vermont College of Medicine, 226 Health Science Research Facility, Burlington, VT USA
| |
Collapse
|
86
|
Meyer KC, Raghu G, Verleden GM, Corris PA, Aurora P, Wilson KC, Brozek J, Glanville AR. An international ISHLT/ATS/ERS clinical practice guideline: diagnosis and management of bronchiolitis obliterans syndrome. Eur Respir J 2014; 44:1479-503. [PMID: 25359357 DOI: 10.1183/09031936.00107514] [Citation(s) in RCA: 397] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bronchiolitis obliterans syndrome (BOS) is a major complication of lung transplantation that is associated with poor survival. The International Society for Heart and Lung Transplantation, American Thoracic Society, and European Respiratory Society convened a committee of international experts to describe and/or provide recommendations for 1) the definition of BOS, 2) the risk factors for developing BOS, 3) the diagnosis of BOS, and 4) the management and prevention of BOS. A pragmatic evidence synthesis was performed to identify all unique citations related to BOS published from 1980 through to March, 2013. The expert committee discussed the available research evidence upon which the updated definition of BOS, identified risk factors and recommendations are based. The committee followed the GRADE (Grading of Recommendation, Assessment, Development and Evaluation) approach to develop specific clinical recommendations. The term BOS should be used to describe a delayed allograft dysfunction with persistent decline in forced expiratory volume in 1 s that is not caused by other known and potentially reversible causes of post-transplant loss of lung function. The committee formulated specific recommendations about the use of systemic corticosteroids, cyclosporine, tacrolimus, azithromycin and about re-transplantation in patients with suspected and confirmed BOS. The diagnosis of BOS requires the careful exclusion of other post-transplant complications that can cause delayed lung allograft dysfunction, and several risk factors have been identified that have a significant association with the onset of BOS. Currently available therapies have not been proven to result in significant benefit in the prevention or treatment of BOS. Adequately designed and executed randomised controlled trials that properly measure and report all patient-important outcomes are needed to identify optimal therapies for established BOS and effective strategies for its prevention.
Collapse
Affiliation(s)
- Keith C Meyer
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ganesh Raghu
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Paul Aurora
- Great Ormond Street Hospital for Children, London, UK
| | | | - Jan Brozek
- McMaster University, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
87
|
Abstract
Lung transplantation has become an important therapeutic option for patients with end-stage organ dysfunction; however, its clinical usefulness has been limited by the relatively early onset of chronic allograft dysfunction and progressive clinical decline. Obliterative bronchiolitis is characterized histologically by luminal fibrosis of the respiratory bronchioles and clinically by bronchiolitis obliterans syndrome (BOS) which is defined by a measured decline in lung function based on forced expiratory volume (FEV1). Since its earliest description, a number of risk factors have been associated with the development of BOS, including acute rejection, lymphocytic bronchiolitis, primary graft dysfunction, infection, donor specific antibodies, and gastroesophageal reflux disease. However, despite this broadened understanding, the pathogenesis underlying BOS remains poorly understood and once begun, there are relatively few treatment options to battle the progressive deterioration in lung function.
Collapse
Affiliation(s)
- Christine M Lin
- 1University of Colorado, Denver - Anschutz Medical Campus, 12700 East 19th Avenue, Room 9470E, Aurora, CO 80045 USA
| | - Martin R Zamora
- 2University of Colorado, Denver - Anschutz Medical Campus, 1635 Aurora Court, Room 7082, Mail Stop F749, Aurora, CO 80045 USA
| |
Collapse
|
88
|
Abstract
Research in pulmonary transplantation is actively evolving in quality and scope to meet the challenges of a growing population of lung allograft recipients. In 2013, research groups leveraged large publicly available datasets in addition to multicenter research networks and single-center studies to make significant contributions to our knowledge and clinical care in the areas of donor use, clinical transplant outcomes, mechanisms of rejection, infectious complications, and chronic allograft dysfunction.
Collapse
Affiliation(s)
- Jamie L Todd
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | | |
Collapse
|
89
|
|
90
|
Shino MY, Weigt SS, Li N, Palchevskiy V, Derhovanessian A, Saggar R, Sayah DM, Gregson AL, Fishbein MC, Ardehali A, Ross DJ, Lynch JP, Elashoff RM, Belperio JA. CXCR3 ligands are associated with the continuum of diffuse alveolar damage to chronic lung allograft dysfunction. Am J Respir Crit Care Med 2013; 188:1117-25. [PMID: 24063316 DOI: 10.1164/rccm.201305-0861oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
RATIONALE After lung transplantation, insults to the allograft generally result in one of four histopathologic patterns of injury: (1) acute rejection, (2) lymphocytic bronchiolitis, (3) organizing pneumonia, and (4) diffuse alveolar damage (DAD). We hypothesized that DAD, the most severe form of acute lung injury, would lead to the highest risk of chronic lung allograft dysfunction (CLAD) and that a type I immune response would mediate this process. OBJECTIVES Determine whether DAD is associated with CLAD and explore the potential role of CXCR3/ligand biology. METHODS Transbronchial biopsies from all lung transplant recipients were reviewed. The association between the four injury patterns and subsequent outcomes were evaluated using proportional hazards models with time-dependent covariates. Bronchoalveolar lavage (BAL) concentrations of the CXCR3 ligands (CXCL9/MIG, CXCL10/IP10, and CXCL11/ITAC) were compared between allograft injury patterns and "healthy" biopsies using linear mixed-effects models. The effect of these chemokine alterations on CLAD risk was assessed using Cox models with serial BAL measurements as time-dependent covariates. MEASUREMENTS AND MAIN RESULTS There were 1,585 biopsies from 441 recipients with 62 episodes of DAD. An episode of DAD was associated with increased risk of CLAD (hazard ratio, 3.0; 95% confidence interval, 1.9-4.7) and death (hazard ratio, 2.3; 95% confidence interval, 1.7-3.0). There were marked elevations in BAL CXCR3 ligand concentrations during DAD. Furthermore, prolonged elevation of these chemokines in serial BAL fluid measurements predicted the development of CLAD. CONCLUSIONS DAD is associated with marked increases in the risk of CLAD and death after lung transplantation. This association may be mediated in part by an aberrant type I immune response involving CXCR3/ligands.
Collapse
Affiliation(s)
- Michael Y Shino
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Saito T, Takahashi H, Kaneda H, Binnie M, Azad S, Sato M, Waddell TK, Cypel M, Liu M, Keshavjee S. Impact of cytokine expression in the pre-implanted donor lung on the development of chronic lung allograft dysfunction subtypes. Am J Transplant 2013; 13:3192-201. [PMID: 24164971 DOI: 10.1111/ajt.12492] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/13/2013] [Accepted: 09/07/2013] [Indexed: 01/25/2023]
Abstract
The long-term success of lung transplantation continues to be challenged by the development of chronic lung allograft dysfunction (CLAD). The purpose of this study was to investigate the relationship between cytokine expression levels in pre-implanted donor lungs and the posttransplant development of CLAD and its subtypes, bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS). Of 109 patients who underwent bilateral lung or heart-lung transplantation and survived for more than 3 months, 50 BOS, 21 RAS and 38 patients with No CLAD were identified by pulmonary function test results. Using donor lung tissue biopsies sampled from each patient, expression levels of IL-6, IL-1β, IL-8, IL-10, interferon-γ and tumor necrosis factor-α mRNA were measured. IL-6 expression levels were significantly higher in pre-implanted lungs of patients that ultimately developed BOS compared to RAS and No CLAD (p = 0.025 and 0.011, respectively). Cox regression analysis demonstrated an association between high IL-6 expression levels and BOS development (hazard ratio = 4.98; 95% confidence interval = 2.42-10.2, p < 0.001). In conclusion, high IL-6 mRNA expression levels in pre-implanted donor lungs were associated with the development of BOS, not RAS. This association further supports the contention that early graft injury impacts on both late graft function and early graft function.
Collapse
Affiliation(s)
- T Saito
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Thoracic and Cardiovascular Surgery, Kansai Medical University, Hirakara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
Primary graft dysfunction (PGD) is a syndrome encompassing a spectrum of mild to severe lung injury that occurs within the first 72 hours after lung transplantation. PGD is characterized by pulmonary edema with diffuse alveolar damage that manifests clinically as progressive hypoxemia with radiographic pulmonary infiltrates. In recent years, new knowledge has been generated on risks and mechanisms of PGD. Following ischemia and reperfusion, inflammatory and immunological injury-repair responses appear to be key controlling mechanisms. In addition, PGD has a significant impact on short- and long-term outcomes; therefore, the choice of donor organ is impacted by this potential adverse consequence. Improved methods of reducing PGD risk and efforts to safely expand the pool are being developed. Ex vivo lung perfusion is a strategy that may improve risk assessment and become a promising platform to implement treatment interventions to prevent PGD. This review details recent updates in the epidemiology, pathophysiology, molecular and genetic biomarkers, and state-of-the-art technical developments affecting PGD.
Collapse
Affiliation(s)
- Yoshikazu Suzuki
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Edward Cantu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jason D Christie
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
93
|
Forgiarini LA, Forgiarini LF, da Rosa DP, Mariano R, Ulbrich JM, Andrade CF. Endobronchial perfluorocarbon administration decreases lung injury in an experimental model of ischemia and reperfusion. J Surg Res 2013; 183:835-40. [PMID: 23434305 DOI: 10.1016/j.jss.2013.01.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 01/12/2013] [Accepted: 01/17/2013] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To verify the effects of liquid endobronchial perfluorocarbon (PFC) administered before reperfusion in an animal model of lung ischemia-reperfusion injury. METHODS Eighteen Wistar rats were subjected to an experimental model of selective left pulmonary artery clamping for 45 min followed by reperfusion for 2 h. The animals were divided into three groups: the ischemia-reperfusion (IR) group, the sham group, and the PFC group. We recorded the hemodynamic parameters, blood gas analysis, and histology. A Western blot assay was used to measure the inducible nitric oxide synthase, caspase 3, and nuclear factor қB (subunit p65) activities. Lipid peroxidation was assessed by the thiobarbituric acid reactive substances assay and the activity of the antioxidant enzyme superoxide dismutase. RESULTS No significant differences were observed in lipid peroxidation among the groups. The superoxide dismutase activity was increased (P < 0.05) in the PFC-treated group. The expressions of nuclear factor қB, inducible nitric oxide synthase, and caspase 3 were significantly lower in the PFC group than in the IR group (P < 0.05). The histologic analysis showed a reduction in lung injuries in the PFC group compared with the sham and IR groups. CONCLUSION The use of endobronchial PFC reduces the inflammatory response, preserves the alveolar structure, and protects the lungs against the hazardous effects of ischemia-reperfusion injuries.
Collapse
Affiliation(s)
- Luiz Alberto Forgiarini
- Postgraduate Program in Pulmonary Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | | | | | | | | | | |
Collapse
|
94
|
Meyer KC, Glanville AR. Bronchiolitis Obliterans Syndrome and Chronic Lung Allograft Dysfunction: Evolving Concepts and Nomenclature. BRONCHIOLITIS OBLITERANS SYNDROME IN LUNG TRANSPLANTATION 2013. [PMCID: PMC7122385 DOI: 10.1007/978-1-4614-7636-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bronchiolitis obliterans syndrome (BOS) eventually occurs in the majority of lung transplant recipients who survive beyond 1 year, can greatly impair quality of life, and is, directly or indirectly, the major cause of delayed allograft dysfunction and recipient death. A number of associated events or conditions are strongly associated with the risk for developing BOS; these include acute rejection, gastroesophageal reflux, infections, and autoimmune reactions that can occur in the setting of alloimmune responses to the lung allograft as recipients are given intense immunosuppression to prevent allograft rejection. The term chronic lung allograft dysfunction (CLAD) is being increasingly used to refer to recipients with late allograft dysfunction that meets the spirometric criteria for the diagnosis of BOS, but clinicians should recognize that such dysfunction can occur for a variety of reasons other than BOS. The recently identified entity of restrictive allograft syndrome, which is now recognized as a relatively distinct phenotype of CLAD, has features that differentiate it from classic obstructive BOS. A number of other entities that can also significantly affect allograft function must also be considered when significant allograft dysfunction is encountered following lung transplantation.
Collapse
|
95
|
Kwon MH, Wong SY, Ardehali A, Laks H, Zhang ZK, Deng MC, Shemin RJ. Primary graft dysfunction does not lead to increased cardiac allograft vasculopathy in surviving patients. J Thorac Cardiovasc Surg 2012; 145:869-73. [PMID: 23083793 DOI: 10.1016/j.jtcvs.2012.09.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 07/16/2012] [Accepted: 09/13/2012] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Early injury is associated with the development of cardiac allograft vasculopathy in heart transplantation. We examined whether adult heart transplant recipients surviving primary graft dysfunction were more susceptible to the development of cardiac allograft vasculopathy than their nonprimary graft dysfunction counterparts. METHODS A total of 857 patients who underwent heart transplantation between January 1994 and December 2008 at our institution were reviewed. Primary graft dysfunction was defined as the need for extracorporeal membrane oxygenation, open chest, or intra-aortic balloon pump placement within 72 hours of transplantation. Cardiac allograft vasculopathy was defined as ≥50% coronary artery stenosis in any vessel. Allograft survival was defined by patient death or need for retransplantation. RESULTS Completed follow-up was available for 32 patients in the primary graft dysfunction group and 701 patients in the nonprimary graft dysfunction group. Mean recipient ages (56 years vs 55 years, respectively; P = .50) and ischemic times (220 minutes vs 208 minutes, respectively; P = .35) were similar. Donor age was significantly higher in the primary graft dysfunction group (38 years vs 32 years, P = .02). Five-year survivals for the primary graft dysfunction and nonprimary graft dysfunction groups were 46.9% versus 78.9% (P < .001). Conditional 5-year survivals in patients surviving the first year were 78.9% and 88.3% for the primary graft dysfunction and nonprimary graft dysfunction groups, respectively (P = .18). Within a 30-day postoperative period, there were more deaths in the primary graft dysfunction group (28.1% vs 2.3%, P < .0001) and more retransplants (6.25% vs 0%, P = .002). Of the patients surviving past 30 days, only 2 (8.7%) of the primary graft dysfunction patients developed cardiac allograft vasculopathy versus 144 (21.0%) in the nonprimary graft dysfunction group (P < .001). CONCLUSIONS Primary graft dysfunction was associated with lower 30-day, 1-year, and 5-year allograft survival rates. Surviving patients, however, did not show increased tendency toward cardiac allograft vasculopathy development.
Collapse
Affiliation(s)
- Murray H Kwon
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
96
|
Hall DJ, Baz M, Daniels MJ, Staples ED, Klodell CT, Moldawer LL, Beaver TM. Immediate postoperative inflammatory response predicts long-term outcome in lung-transplant recipients. Interact Cardiovasc Thorac Surg 2012; 15:603-7. [PMID: 22815323 DOI: 10.1093/icvts/ivs330] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Although lung transplantation is an accepted therapy for end-stage disease, recipient outcomes continue to be hindered by early primary graft dysfunction (PGD) as well as late rejection and bronchiolitis obliterans syndrome (BOS). We have previously shown that the pro-inflammatory cytokine response following transplantation correlates with the severity of PGD. We hypothesized that lung-transplant recipients with an increased inflammatory response immediately following surgery would also have a greater incidence of unfavorable long-term outcomes including rejection, BOS and ultimately death. METHODS A retrospective study of lung-transplant recipients (n = 19) for whom serial blood sampling of cytokines was performed for 24 h following transplantation between March 2002 and June 2003 at a single institution. Long-term follow-up was examined for rejection, BOS and survival. RESULTS Thirteen single and six bilateral lung recipients were examined. Eleven (58%) developed BOS and eight (42%) did not. Subgroup analysis revealed an association between elevated IL-6 concentrations 4 h after reperfusion of the allograft and development of BOS (P = 0.068). The correlation between IL-6 and survival time was found to be significant (corr = -0.46, P = 0.047), indicating that higher IL-6 response had shorter survival following transplantation. CONCLUSIONS An elevation in interleukin (IL)-6 concentration immediately following lung transplantation is associated with a trend towards development of bronchiolitis obliterans, rejection and significantly decreased survival time. Further studies are warranted to confirm the correlation between the immediate inflammatory response, PGD and BOS. Identification of patients at risk for BOS based on the cytokine response after surgery may allow for early intervention.
Collapse
Affiliation(s)
- David J Hall
- College of Medicine, University of Florida, Gainesville, FL 32610-0286, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Sarma NJ, Tiriveedhi V, Angaswamy N, Mohanakumar T. Role of antibodies to self-antigens in chronic allograft rejection: potential mechanism and therapeutic implications. Hum Immunol 2012; 73:1275-81. [PMID: 22789626 DOI: 10.1016/j.humimm.2012.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/21/2012] [Accepted: 06/29/2012] [Indexed: 02/07/2023]
Abstract
Significant progress has been made in preventing acute allograft rejection following solid organ transplantation resulting in improved allograft survival. However, long term function still remains disappointing primarily due to chronic allograft rejection. Alloimmune responses primarily defined by the development of antibodies (Abs) to donor mismatched major histocompatibility antigens during the post-transplantation period have been strongly correlated to the development of chronic rejection. In addition, recent studies have demonstrated an important role for autoimmunity including the development of Abs to organ specific self-antigens in the pathogenesis of chronic allograft rejection. Based on this, a new paradigm has evolved indicating a possible cross-talk between the alloimmune responses and autoimmunity leading to chronic rejection. In this review, we will discuss the emerging concept for the role of cellular and humoral immune responses to self-antigens in the immunopathogenesis of chronic allograft rejection which has the potential to develop new strategies for the prevention and/or treatment of chronic rejection.
Collapse
Affiliation(s)
- Nayan J Sarma
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States
| | | | | | | |
Collapse
|
98
|
Gade J, Greisen G, Larsen IK, Bibby BM, Olsen PS. Tissue hypoxaemia causes oedema, inflammation and fibrosis in porcine bronchial transsection. SCAND CARDIOVASC J 2012; 46:286-94. [PMID: 22607392 DOI: 10.3109/14017431.2012.695086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Bronchial artery revascularization in lung transplantation is disputed. This study examined the physiological consequences of porcine bronchial transsection and reanastomosis with and without bronchial artery blood supply with relation to lung transplantation. DESIGN Translational, controlled animal study. Twelve pigs were operated through a left lateral thoracotomy. The left bronchus was transsected and reanastomosed. In the control group (n = 6), the bronchial arteries were preserved and in the study group (n = 6) they were severed. Bronchial mucosa blood flow (BMBF) was measured with laser-Doppler velocimetry and bronchial mucosa haemoglobin saturation and concentration with diffuse reflectance spectrophotometry. Measurements were made preoperatively, postoperatively and after 1 week. RESULTS In the study group, left postoperative BMBF was significantly lower than preoperatively (115 vs. 210 PU/s, p = 0.0001) and lower than in the control group (115 vs. 205 PU/s, p = 0.002). Repeated measurement ANOVA showed a significant treatment effect depending on time (p = 0.0034). The left mucosal haemoglobin saturation in the study group was significantly reduced postoperatively, 92% versus 61%, with a treatment effect depending on time (p = 0.0080). The reduction in left/right ratio of the mucosal haemoglobin concentration 1 week postoperatively in the study group was insignificant. CONCLUSION Bronchial transsection and reanastomosis without bronchial artery blood supply was followed by significant decrease in mucosal blood flow and saturation postoperatively, and also in tissue haemoglobin concentration at section, and provides a physiologic explanation of histological changes.
Collapse
Affiliation(s)
- John Gade
- Department of Cardiothoracic Surgery RT, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | | | | | | | | |
Collapse
|
99
|
Cumulative exposure to CD8+ granzyme Bhi T cells is associated with reduced lung function early after lung transplantation. Transplant Proc 2012; 43:3892-8. [PMID: 22172867 DOI: 10.1016/j.transproceed.2011.09.072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 09/16/2011] [Indexed: 11/20/2022]
Abstract
Outcomes following lung transplant remain suboptimal. This is attributable to variable posttransplant recovery of lung function, and inconsistent degrees of lung function loss after peak function is reached. Granzyme B is elevated in the blood and bronchoalveolar lavage (BAL) in acute rejection. We hypothesized that persistent exposure to T cells high in granzyme B would negatively correlate with lung function. We investigated cumulative exposure measured as the area-under-the-curve (AUC) of CD8+ T cell granzyme Bhi cells in the first year posttransplant in both BAL and blood in 24 transplant recipients. We assessed the correlation between cumulative 1-year exposure and FEV1 slope. There was a negative correlation between 1-year exposure and FEV1 slope within the first year (r=-0.63; P=.001). This relationship persisted even when adjusted for transplant type, gender, age, rejection, and indication for transplantation. In contrast, no relationship was seen with the 1-year AUC and lung function after 1 year posttransplant. In contrast to the BAL granzyme Bhi levels, granzyme Bhi levels from the blood showed no relationship with lung function. These findings suggest that CD8+ T-cell-driven factors are responsible for early improvements in lung function after transplantation.
Collapse
|
100
|
Benson HL, Suzuki H, Lott J, Fisher AJ, Walline C, Heidler KM, Brutkiewicz R, Blum JS, Wilkes DS. Donor lung derived myeloid and plasmacytoid dendritic cells differentially regulate T cell proliferation and cytokine production. Respir Res 2012; 13:25. [PMID: 22433165 PMCID: PMC3352265 DOI: 10.1186/1465-9921-13-25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/20/2012] [Indexed: 11/17/2022] Open
Abstract
Background Direct allorecognition, i.e., donor lung-derived dendritic cells (DCs) stimulating recipient-derived T lymphocytes, is believed to be the key mechanism of lung allograft rejection. Myeloid (cDCs) and plasmacytoid (pDCs) are believed to have differential effects on T cell activation. However, the roles of each DC type on T cell activation and rejection pathology post lung transplantation are unknown. Methods Using transgenic mice and antibody depletion techniques, either or both cell types were depleted in lungs of donor BALB/c mice (H-2d) prior to transplanting into C57BL/6 mice (H-2b), followed by an assessment of rejection pathology, and pDC or cDC-induced proliferation and cytokine production in C57BL/6-derived mediastinal lymph node T cells (CD3+). Results Depleting either DC type had modest effect on rejection pathology and T cell proliferation. In contrast, T cells from mice that received grafts depleted of both DCs did not proliferate and this was associated with significantly reduced acute rejection scores compared to all other groups. cDCs were potent inducers of IFNγ, whereas both cDCs and pDCs induced IL-10. Both cell types had variable effects on IL-17A production. Conclusion Collectively, the data show that direct allorecognition by donor lung pDCs and cDCs have differential effects on T cell proliferation and cytokine production. Depletion of both donor lung cDC and pDC could prevent the severity of acute rejection episodes.
Collapse
Affiliation(s)
- Heather L Benson
- Department of Medicine, Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|