Your browser doesn't support javascript.
loading
Systems prediction of chronic lung allograft dysfunction: Results and perspectives from the Cohort of Lung Transplantation and Systems prediction of Chronic Lung Allograft Dysfunction cohorts.
Pison, Christophe; Tissot, Adrien; Bernasconi, Eric; Royer, Pierre-Joseph; Roux, Antoine; Koutsokera, Angela; Coiffard, Benjamin; Renaud-Picard, Benjamin; Le Pavec, Jérôme; Mordant, Pierre; Demant, Xavier; Villeneuve, Thomas; Mornex, Jean-Francois; Nemska, Simona; Frossard, Nelly; Brugière, Olivier; Siroux, Valérie; Marsland, Benjamin J; Foureau, Aurore; Botturi, Karine; Durand, Eugenie; Pellet, Johann; Danger, Richard; Auffray, Charles; Brouard, Sophie; Nicod, Laurent; Magnan, Antoine.
Afiliação
  • Pison C; Service Hospitalier Universitaire de Pneumologie Physiologie, Pôle Thorax et Vaisseaux, Fédération Grenoble Transplantation, CHU Grenoble Alpes, Grenoble, France.
  • Tissot A; Université Grenoble Alpes, INSERM 1055, Grenoble, France.
  • Bernasconi E; Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France.
  • Royer PJ; CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France.
  • Roux A; Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse.
  • Koutsokera A; CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France.
  • Coiffard B; Service de Pneumologie, Hôpital Foch, Suresnes, France.
  • Renaud-Picard B; Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, INRAE, Jouy-en-Josas, France.
  • Le Pavec J; Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse.
  • Mordant P; Service de Pneumologie et de Transplantation Pulmonaire, APHM, Hôpital Nord, Aix Marseille Univ, Marseille, France.
  • Demant X; Service de Pneumologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
  • Villeneuve T; Inserm UMR 1260, Regenerative Nanomedicine, Université de Strasbourg, Strasbourg, France.
  • Mornex JF; Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardiopulmonaire, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France.
  • Nemska S; Service de Chirurgie Vasculaire, Thoracique et Transplantation Pulmonaire, Hôpital Bichat, AP-HP, INSERM U1152, Université Paris Cité, Paris, France.
  • Frossard N; Service de Pneumologie et Transplantation Pulmonaire, CHU de Bordeaux, Bordeaux, France.
  • Brugière O; Service de Pneumologie, CHU de Toulouse, Université Toulouse III-Paul Sabatier, Toulouse, France.
  • Siroux V; Université de Lyon, Université Lyon 1, PSL, EPHE, INRAE, IVPC, Lyon, France.
  • Marsland BJ; Hospices Civils de Lyon, GHE, Service de Pneumologie, RESPIFIL, Orphalung, Inserm CIC, Lyon, France.
  • Foureau A; UMR 7200 - Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, CNRS-Université de Strasbourg, Illkirch, France.
  • Botturi K; UMR 7200 - Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, CNRS-Université de Strasbourg, Illkirch, France.
  • Durand E; Service de Pneumologie, Hôpital Foch, Suresnes, France.
  • Pellet J; Laboratoire d'Immunologie de la Transplantation, Hôpital Saint-Louis, CEA/DRF/Institut de Biologie François Jacob, Unité INSERM 1152, Université Paris Diderot, USPC, Paris, France.
  • Danger R; Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences (IAB), Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.
  • Auffray C; Unité de Transplantation Pulmonaire, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Lausanne, Suisse.
  • Brouard S; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.
  • Nicod L; Service de Pneumologie, Institut du Thorax, CHU Nantes, Nantes, France.
  • Magnan A; CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France.
Front Med (Lausanne) ; 10: 1126697, 2023.
Article em En | MEDLINE | ID: mdl-36968829
ABSTRACT

Background:

Chronic lung allograft dysfunction (CLAD) is the leading cause of poor long-term survival after lung transplantation (LT). Systems prediction of Chronic Lung Allograft Dysfunction (SysCLAD) aimed to predict CLAD.

Methods:

To predict CLAD, we investigated the clinicome of patients with LT; the exposome through assessment of airway microbiota in bronchoalveolar lavage cells and air pollution studies; the immunome with works on activation of dendritic cells, the role of T cells to promote the secretion of matrix metalloproteinase-9, and subpopulations of T and B cells; genome polymorphisms; blood transcriptome; plasma proteome studies and assessment of MSK1 expression.

Results:

Clinicome the best multivariate logistic regression analysis model for early-onset CLAD in 422 LT eligible patients generated a ROC curve with an area under the curve of 0.77. Exposome chronic exposure to air pollutants appears deleterious on lung function levels in LT recipients (LTRs), might be modified by macrolides, and increases mortality. Our findings established a link between the lung microbial ecosystem, human lung function, and clinical stability post-transplant. Immunome a decreased expression of CLEC1A in human lung transplants is predictive of the development of chronic rejection and associated with a higher level of interleukin 17A; Immune cells support airway remodeling through the production of plasma MMP-9 levels, a potential predictive biomarker of CLAD. Blood CD9-expressing B cells appear to favor the maintenance of long-term stable graft function and are a potential new predictive biomarker of BOS-free survival. An early increase of blood CD4 + CD57 + ILT2+ T cells after LT may be associated with CLAD onset. Genome Donor Club cell secretory protein G38A polymorphism is associated with a decreased risk of severe primary graft dysfunction after LT. Transcriptome blood POU class 2 associating factor 1, T-cell leukemia/lymphoma domain, and B cell lymphocytes, were validated as predictive biomarkers of CLAD phenotypes more than 6 months before diagnosis. Proteome blood A2MG is an independent predictor of CLAD, and MSK1 kinase overexpression is either a marker or a potential therapeutic target in CLAD.

Conclusion:

Systems prediction of Chronic Lung Allograft Dysfunction generated multiple fingerprints that enabled the development of predictors of CLAD. These results open the way to the integration of these fingerprints into a predictive handprint.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Med (Lausanne) Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Med (Lausanne) Ano de publicação: 2023 Tipo de documento: Article