Complement inhibition ameliorates blast-induced acute lung injury in rats: Potential role of complement in intracellular HMGB1-mediated inflammation

PLoS One. 2018 Aug 22;13(8):e0202594. doi: 10.1371/journal.pone.0202594. eCollection 2018.

Abstract

Background and objective: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action.

Methods: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500μg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR.

Results: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs.

Conclusions: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acute Lung Injury / drug therapy*
  • Acute Lung Injury / genetics
  • Acute Lung Injury / physiopathology
  • Animals
  • Blast Injuries / drug therapy*
  • Blast Injuries / genetics
  • Blast Injuries / pathology
  • CD55 Antigens / administration & dosage*
  • Complement Activation / drug effects
  • Complement C3a / antagonists & inhibitors
  • Disease Models, Animal
  • HMGB1 Protein / genetics*
  • Humans
  • Inflammation / drug therapy*
  • Inflammation / genetics
  • Inflammation / physiopathology
  • Lung / drug effects
  • Lung / metabolism
  • Lung / physiopathology
  • NF-kappa B / genetics
  • Pressure / adverse effects
  • Rats
  • Rats, Sprague-Dawley

Substances

  • CD55 Antigens
  • HMGB1 Protein
  • Hbp1 protein, rat
  • NF-kappa B
  • Complement C3a

Grants and funding

This study was supported by the Defense Medical Research and Development Program for supporting this work (grant# D61_I_10_J6_82). http://cdmrp.army.mil/dmrdp/ The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.