RT Journal Article SR Electronic T1 A type I IFN, prothrombotic hyperinflammatory neutrophil signature is distinct for COVID-19 ARDS JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2020.09.15.20195305 DO 10.1101/2020.09.15.20195305 A1 Leila Reyes A1 Manuel A. Sanchez-Garcia A1 Tyler Morrison A1 Andrew J.M. Howden A1 Emily R. Watts A1 Simone Arienti A1 Pranvera Sadiku A1 Patricia Coelho A1 Ananda S. Mirchandani A1 Ailiang Zhang A1 David Hope A1 Sarah K. Clark A1 Jo Singleton A1 Shonna Johnston A1 Robert Grecian A1 Azin Poon A1 Sarah McNamara A1 Isla Harper A1 Max Head Fourman A1 Alejandro J. Brenes A1 Shalini Pathak A1 Amy Lloyd A1 Giovanny Rodriguez Blanco A1 Alex von Kriegsheim A1 Bart Ghesquiere A1 Wesley Vermaelen A1 Camila T. Cologna A1 Kevin Dhaliwal A1 Nik Hirani A1 David H. Dockrell A1 Moira K. B. Whyte A1 David Griffith A1 Doreen A. Cantrell A1 Sarah R. Walmsley YR 2021 UL http://medrxiv.org/content/early/2021/01/28/2020.09.15.20195305.abstract AB Acute respiratory distress syndrome (ARDS) is a severe critical condition with a high mortality that is currently in focus given that it is associated with mortality caused by coronavirus induced disease 2019 (COVID-19). Neutrophils play a key role in the lung injury characteristic of non-COVID-19 ARDS and there is also accumulating evidence of neutrophil mediated lung injury in patients who succumb to infection with SARS-CoV-2. We undertook a functional proteomic and metabolomic survey of circulating neutrophil populations, comparing patients with COVID-19 ARDS and non-COVID-19 ARDS to understand the molecular basis of neutrophil dysregulation. Expansion of the circulating neutrophil compartment and the presence of activated low and normal density mature and immature neutrophil populations occurs in ARDS, irrespective of cause. Release of neutrophil granule proteins, neutrophil activation of the clotting cascade and upregulation of the Mac-1 platelet binding complex with formation of neutrophil platelet aggregates is exaggerated in COVID-19 ARDS. Importantly, activation of components of the neutrophil type I interferon responses is seen in ARDS following infection with SARS-CoV-2, with associated rewiring of neutrophil metabolism, and the upregulation of antigen processing and presentation. Whilst dexamethasone treatment constricts the immature low density neutrophil population it does not impact upon prothrombotic hyperinflammatory neutrophil signatures.Summary Given the crucial role of neutrophils in ARDS and the evidence of a disordered myeloid response observed in COVID-19 patients, our work maps the molecular basis for neutrophil reprogramming in the distinct clinical entities of COVID-19 and non-COVID-19 ARDS.Conclusions This work maps the molecular basis for neutrophil reprogramming in the distinct clinical entities of COVID-19 and non-COVID-19 ARDS.Competing Interest StatementThe authors have declared no competing interest.Funding StatementThis research was supported by a Wellcome Trust Senior Clinical Fellowship award (209220) and a CRUK cancer immunology project award (C62207/A24495) to S.R.W, Wellcome Clinical training Fellowship awards to T.M. (214383/Z/18/Z) and E.R.W (108717/Z/15/Z), a Wellcome Trust Post-doctoral Training Clinical Fellowship awarded to A.S.M (110086), a Medical Research Foundation PhD Studentship to S.A., UKRI/NIHR funding through the UK Coronavirus Immunology Consortium (UK-CIC) and a CSO grant (COV/DUN/20/01) to D.A.C, and a LifeArc STOPCOVID award to A.P and S.M.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Human peripheral venous blood was taken from healthy volunteers with written informed consent obtained from all participants prior to sample collection as approved by the University of Edinburgh Centre for Inflammation Research Blood Resource Management Committee (AMREC 15-HV-013). The collection of peripheral venous blood from male or female patients diagnosed with COVID-19 and/or presenting with ARDS was approved by Scotland A Research Ethics Committee. Patient recruitment took place from April 2020 through August 2020 from The Royal Infirmary of Edinburgh, Scotland, UK through the ARDS Neut (20/SS/0002) and CASCADE (20/SS/0052) Study, with informed consent obtained by proxy.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesRaw mass spectrometry data files and Spectronaut analysis files will be available to download from the ProteomeXchange data repository (http://proteomecentral.proteomexchange.org/cgi/GetDataset) at the time of publication.