@article{Reyes_L-2021_26427, title = {A type I IFN, prothrombotic hyperinflammatory neutrophil signature is distinct for COVID-19 ARDS­­­}, author = {Reyes, L. and Sanchez-Garcia, M. and Morrison, T. and Howden, A. and Watts, E. and Arienti, S. and Sadiku, P. and Coelho, P. and Mirchandani, A. and Zhang, A. and Hope, D. and Clark, S. and Singleton, J. and Johnston, S. and Grecian, R. and Poon, A. and McNamara, S. and Harper, I. and Fourman, M. and Brenes Murillo, A. and Pathak, S. and Lloyd, A. and Rodriguez Blanco, G. and von Kriegsheim, A. and Ghesquiere, B. and Vermaelen, W. and Cologna, C. and Dhaliwal, K. and Hirani, N. and Dockrell, D. and Whyte, M. and Griffith, D. and Cantrell, D. and Walmsley, S.}, month = {feb}, year = {2021}, abstract = {Background: 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 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 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Methods: 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. Results: 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. Conclusions: Given the crucial role of neutrophils in ARDS and the evidence of a disordered myeloid response observed in COVID-19 patients, this work maps the molecular basis for neutrophil reprogramming in the distinct clinical entities of COVID-19 and non-COVID-19 ARDS.}, volume = {6}, journal = {Wellcome Open Research}, publisher = {Wellcome Open Research}, url = {https://doi.org/10.12688/wellcomeopenres.16584.1}, }