Release of neutrophil extracellular traps by neutrophils stimulated with antiphospholipid antibodies: a newly identified mechanism of thrombosis in the …

S Yalavarthi, TJ Gould, AN Rao, LF Mazza… - Arthritis & …, 2015 - Wiley Online Library
S Yalavarthi, TJ Gould, AN Rao, LF Mazza, AE Morris, C Núñez‐Álvarez…
Arthritis & rheumatology, 2015Wiley Online Library
Objective Antiphospholipid antibodies (aPL), especially those targeting β2‐glycoprotein I
(β2GPI), are well known to activate endothelial cells, monocytes, and platelets, with
prothrombotic implications. In contrast, the interaction of aPL with neutrophils has not been
extensively studied. Neutrophil extracellular traps (NETs) have recently been recognized as
an important activator of the coagulation cascade, as well as an integral component of
arterial and venous thrombi. This study was undertaken to determine whether aPL activate …
Objective
Antiphospholipid antibodies (aPL), especially those targeting β2‐glycoprotein I (β2GPI), are well known to activate endothelial cells, monocytes, and platelets, with prothrombotic implications. In contrast, the interaction of aPL with neutrophils has not been extensively studied. Neutrophil extracellular traps (NETs) have recently been recognized as an important activator of the coagulation cascade, as well as an integral component of arterial and venous thrombi. This study was undertaken to determine whether aPL activate neutrophils to release NETs, thereby predisposing to the arterial and venous thrombosis inherent in the antiphospholipid syndrome (APS).
Methods
Neutrophils, sera, and plasma were prepared from patients with primary APS (n = 52) or from healthy volunteers and characterized. No patient had concomitant systemic lupus erythematosus.
Results
Sera and plasma from patients with primary APS had elevated levels of both cell‐free DNA and NETs, as compared to healthy volunteers. Freshly isolated neutrophils from patients with APS were predisposed to high levels of spontaneous NET release. Further, APS patient sera, as well as IgG purified from APS patients, stimulated NET release from control neutrophils. Human aPL monoclonal antibodies, especially those targeting β2GPI, also enhanced NET release. The induction of APS NETs was abrogated with inhibitors of reactive oxygen species formation and Toll‐like receptor 4 signaling. Highlighting the potential clinical relevance of these findings, APS NETs promoted thrombin generation.
Conclusion
Our findings indicate that NET release warrants further investigation as a novel therapeutic target in APS.
Wiley Online Library