Recombinant human DNase-I improves acute respiratory distress syndrome via neutrophil extracellular trap degradation

Abbas Jarrahi, Hesam Khodadadi, Nicholas S. Moore, Yujiao Lu, Mohamed E. Awad, Evila L. Salles, Kumar Vaibhav, Babak Baban, Krishnan M. Dhandapani

Research output: Contribution to journalArticlepeer-review

4 Scopus citations

Abstract

Background: Respiratory failure is the primary cause of death in patients with COVID-19, whereas coagulopathy is associated with excessive inflammation and multiorgan failure. Neutrophil extracellular traps (NETs) may exacerbate inflammation and provide a scaffold for thrombus formation. Objectives: The goal of this study was to determine whether degradation of NETs by recombinant human DNase-I (rhDNase), a safe, Food and Drug Administration–approved drug, reduces excessive inflammation, reverses aberrant coagulation, and improves pulmonary perfusion after experimental acute respiratory distress syndrome (ARDS). Methods: Intranasal poly(I:C), a synthetic double-stranded RNA, was administered to adult mice for 3 consecutive days to simulate a viral infection, and these subjects were randomized to treatment arms, which received either an intravenous placebo or rhDNase. The effects of rhDNase on immune activation, platelet aggregation, and coagulation were assessed in mice and donor human blood. Results: NETs were observed in bronchoalveolar lavage fluid and within regions of hypoxic lung tissue after experimental ARDS. The administration of rhDNase mitigated peribronchiolar, perivascular, and interstitial inflammation induced by poly(I:C). In parallel, rhDNase degraded NETs, attenuated platelet-NET aggregates, reduced platelet activation, and normalized the clotting time to improve regional perfusion, as observed using gross morphology, histology, and microcomputed tomographic imaging in mice. Similarly, rhDNase reduced NETs and attenuated platelet activation in human blood. Conclusion: NETs exacerbate inflammation and promote aberrant coagulation by providing a scaffold for aggregated platelets after experimental ARDS. Intravenous administration of rhDNase degrades NETs and attenuates coagulopathy in ARDS, providing a promising translational approach to improve pulmonary structure and function after ARDS.

Original languageEnglish (US)
Pages (from-to)2473-2484
Number of pages12
JournalJournal of Thrombosis and Haemostasis
Volume21
Issue number9
DOIs
StatePublished - Sep 2023

Keywords

  • coagulation
  • immunothrombosis
  • inflammation
  • platelets
  • thrombosis

ASJC Scopus subject areas

  • Hematology

Fingerprint

Dive into the research topics of 'Recombinant human DNase-I improves acute respiratory distress syndrome via neutrophil extracellular trap degradation'. Together they form a unique fingerprint.

Cite this