Neutrophil extracellular traps promote tPA-induced brain hemorrhage via cGAS in mice with stroke

R Wang, Y Zhu, Z Liu, L Chang, X Bai… - Blood, The Journal …, 2021 - ashpublications.org
R Wang, Y Zhu, Z Liu, L Chang, X Bai, L Kang, Y Cao, X Yang, H Yu, MJ Shi, Y Hu, W Fan…
Blood, The Journal of the American Society of Hematology, 2021ashpublications.org
Intracerebral hemorrhage associated with thrombolytic therapy with tissue plasminogen
activator (tPA) in acute ischemic stroke continues to present a major clinical problem. Here,
we report that infusion of tPA resulted in a significant increase in markers of neutrophil
extracellular traps (NETs) in the ischemic cortex and plasma of mice subjected to
photothrombotic middle cerebral artery occlusion. Peptidylarginine deiminase 4 (PAD4), a
critical enzyme for NET formation, is also significantly upregulated in the ischemic brains of …
Abstract
Intracerebral hemorrhage associated with thrombolytic therapy with tissue plasminogen activator (tPA) in acute ischemic stroke continues to present a major clinical problem. Here, we report that infusion of tPA resulted in a significant increase in markers of neutrophil extracellular traps (NETs) in the ischemic cortex and plasma of mice subjected to photothrombotic middle cerebral artery occlusion. Peptidylarginine deiminase 4 (PAD4), a critical enzyme for NET formation, is also significantly upregulated in the ischemic brains of tPA-treated mice. Blood–brain barrier (BBB) disruption after ischemic challenge in an in vitro model of BBB was exacerbated after exposure to NETs. Importantly, disruption of NETs by DNase I or inhibition of NET production by PAD4 deficiency restored tPA-induced loss of BBB integrity and consequently decreased tPA-associated brain hemorrhage after ischemic stroke. Furthermore, either DNase I or PAD4 deficiency reversed tPA-mediated upregulation of the DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS). Administration of cGAMP after stroke abolished DNase I–mediated downregulation of the STING pathway and type 1 interferon production and blocked the antihemorrhagic effect of DNase I in tPA-treated mice. We also show that tPA-associated brain hemorrhage after ischemic stroke was significantly reduced in cGas−/− mice. Collectively, these findings demonstrate that NETs significantly contribute to tPA-induced BBB breakdown in the ischemic brain and suggest that targeting NETs or cGAS may ameliorate thrombolytic therapy for ischemic stroke by reducing tPA-associated hemorrhage.
ashpublications.org