Aspirin-triggered 15-epi-lipoxin A4 regulates neutrophil-platelet aggregation and attenuates acute lung injury in mice

G Ortiz-Muņoz, B Mallavia, A Bins… - Blood, The Journal …, 2014 - ashpublications.org
G Ortiz-Muņoz, B Mallavia, A Bins, M Headley, MF Krummel, MR Looney
Blood, The Journal of the American Society of Hematology, 2014ashpublications.org
Evidence is emerging that platelets are major contributors to innate immune responses in
conditions such as acute lung injury (ALI). Platelets form heterotypic aggregates with
neutrophils, and we hypothesized that lipoxin mediators regulate formation of neutrophil-
platelet aggregates (NPA) and that NPA significantly contribute to ALI. Lipopolysaccharide
(LPS)-induced lung injury was accompanied by platelet sequestration, activation, intra-
alveolar accumulation, and NPA formation within both blood and alveolar compartments …
Abstract
Evidence is emerging that platelets are major contributors to innate immune responses in conditions such as acute lung injury (ALI). Platelets form heterotypic aggregates with neutrophils, and we hypothesized that lipoxin mediators regulate formation of neutrophil-platelet aggregates (NPA) and that NPA significantly contribute to ALI. Lipopolysaccharide (LPS)-induced lung injury was accompanied by platelet sequestration, activation, intra-alveolar accumulation, and NPA formation within both blood and alveolar compartments. Using lung intravital microscopy, we observed the dynamic formation of NPA during physiologic conditions, which sharply increased with ALI. Aspirin (ASA) treatment significantly reduced lung platelet sequestration and activation, NPA formation, and lung injury. ASA treatment increased levels of ASA-triggered lipoxin (ATL; 15-epi-lipoxin A4), and blocking the lipoxin A4 receptor (ALX) with a peptide antagonist (Boc2) or using ALX knockouts (Fpr2/3−/−) reversed this protection. LPS increased NPA formation in vitro, which was reduced by ATL, and engagement of ALX by ATL on both neutrophils and platelets was necessary to prevent aggregation. In a model of transfusion-related acute lung injury (TRALI), Boc2 also reversed ASA protection, and treatment with ATL in both LPS and TRALI models protected from ALI. We conclude that ATL regulates neutrophil-platelet aggregation and that platelet-neutrophil interactions are a therapeutic target in lung injury.
ashpublications.org