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

Ranran Wang, Yuanbo Zhu, Zhongwang Liu, Luping Chang, Xiaofei Bai, Lijing Kang, Yongliang Cao, Xing Yang, Huilin Yu, Mei-Juan Shi, Yue Hu, Wenying Fan, Bing-Qiao Zhao, Ranran Wang, Yuanbo Zhu, Zhongwang Liu, Luping Chang, Xiaofei Bai, Lijing Kang, Yongliang Cao, Xing Yang, Huilin Yu, Mei-Juan Shi, Yue Hu, Wenying Fan, Bing-Qiao Zhao

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.

Conflict of interest statement

Conflict-of-interest disclosure: The authors declare no competing financial interests.

© 2021 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
tPA activates neutrophils to release NETs after ischemic stroke. (A-B) Representative immunoblots and quantitative determinations of the amount of neutrophils in the ischemic cortex at 24 hours after stroke in mice treated with vehicle or tPA compared with mice undergoing sham surgery (n = 5). (C) Quantification of the numbers of infiltrating neutrophils in the ischemic cortex at 24 hours after stroke (n = 6). (D) Quantification of MPO activity in the ischemic brain (n = 6). (E-F) Representative immunoblots and quantitative determinations of H3Cit levels in the ischemic cortex (n = 5). (G) Representative confocal images of H3Cit+ neutrophils in the peri-infarct cortex. Scale bar, 50 μm. (H) Quantification of plasma DNA (n = 6). (I) Representative images of isolated peripheral blood neutrophils from mice undergoing sham surgery and ischemic mice treated with vehicle or tPA. Neutrophils were incubated with LPS for 2.5 hours. DNA was stained with Hoechst 33342 (blue), and neutrophils were stained with H3Cit (green). Arrows indicate NETs. Scale bar, 50 μm. (J-K) Quantification of the percentage of H3Cit+ neutrophils and NETs in unstimulated (US) and LPS-stimulated peripheral blood neutrophils (n = 8). (L-M) Quantification of the percentage of H3Cit+ neutrophils and NETs in peripheral blood neutrophils isolated from naïve mice, mice undergoing sham surgery, or ischemic mice (n = 5-8). Neutrophils from naive mice or mice undergoing sham surgery were treated with vehicle or 100 μg/mL of tPA. Neutrophils from ischemic mice were treated with vehicle or 25, 50, or 100 μg/mL of tPA. (N) Representative immunoblots of PAD4 expression in neutrophils isolated from ischemic mice. Neutrophils were treated with vehicle or 100 μg/mL of tPA. (O-P) Quantification of the percentage of H3Cit+ neutrophils and NETs in neutrophils isolated from ischemic mice (n = 8). Neutrophils were treated with vehicle, 100 μg/mL of tPA, or tPA in combination with the PAD inhibitor Cl-amidine. (Q) Representative immunoblots of lipoprotein receptor–related protein 1 (LRP-1) expression in neutrophils isolated from ischemic mice. Neutrophils were treated with vehicle or 100 μg/mL of tPA. (R) Representative immunoblots of PAD4 expression in neutrophils isolated from ischemic mice. Neutrophils were treated with 100 μg/mL of tPA or tPA in combination with the LRP antagonist RAP. (S-T) Quantification of the percentage of H3Cit+ neutrophils and NETs in neutrophils isolated from ischemic mice (n = 8). Neutrophils were treated with vehicle, 100 μg/mL of tPA, or tPA in combination with the LRP antagonist RAP. Values are means ± standard deviation. *P < .05. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.
Figure 2.
Figure 2.
LRP-1 mediates the effects of tPA on neutrophil recruitment and NET formation after ischemic stroke. (A-B) Representative immunoblots and quantitative determinations of LRP-1 expression in the ischemic cortex at 24 hours after stroke in mice treated with vehicle or tPA compared with mice undergoing sham surgery (n = 5). (C) Representative confocal images of LRP-1 immunostaining in the ischemic cortex. DNA was stained with 4′,6-diamidino-2-phenylindole (blue). Independent experiments are repeated ≥3 times. Scale bar, 40 μm. (D) Quantification of MPO activity in the ischemic brain (n = 6). (E) Representative immunoblots of H3Cit in the ischemic cortex. (F) Quantification of MPO activity in the ischemic brain (n = 6). (G) Representative immunoblots of H3Cit in the ischemic cortex. (H) Representative confocal images of fibrin intravascular deposits (green) and CD31+ microvessels (red) in the infarct areas at 24 hours. Scale bar, 10 μm. (I) Quantification of fibrin intravascular deposits for each group (n = 6). Values are means ± standard deviation. *P < .05. NS, not significant; TXA, tranexamic acid.
Figure 3.
Figure 3.
DNase I treatment reduces tPA-mediated BBB breakdown and cerebral hemorrhage after ischemic stroke. (A) Permeability coefficient (P) of human brain endothelial monolayers to 40 KDa of FITC-dextran at 0 to 6 hours after normoxia or OGD with or without NETs (1.5 μg/mL). (B) Representative immunoblots of H3Cit levels in the ischemic cortex at 24 hours after stroke in mice treated with vehicle, tPA, or tPA in combination with DNase I. (C) Quantification of the numbers of H3Cit+ neutrophils in the ischemic cortex (n = 6). (D) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show Evans blue extravasation 24 hours after stroke in mice treated with vehicle, tPA, tPA in combination with DNase I, or DNase I alone. (E) Quantification of Evans blue fluorescence intensity for each group (n = 8). (F-G) Representative immunoblots and quantification of immunoglobulin G (IgG) levels in capillary-depleted brain tissue at 24 hours after stroke (n = 5). (H) Representative immunoblots of the tight junction proteins zonula occludens-1 (ZO-1), claudin-5, and occludin and the adherens junction protein vascular endothelial–cadherin (VE-cadherin) in isolated brain microvessels. (I) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show cerebral hemorrhage 24 hours after stroke in mice treated with vehicle, tPA, tPA in combination with DNase I, or DNase I alone. (J) Quantification of cerebral hemorrhage by spectrophotometric hemoglobin assay (n = 8). (K-M) DNase I treatment improved neurological functions in forelimb force test and beam walking test (n = 10). Values are means ± standard deviation. *P < .05 vs control group (A), #P < .05 vs OGD group (A), *P < .05 (C,E,G,J-M).
Figure 4.
Figure 4.
NETs and PAD4 contribute to tPA-mediated BBB disruption and cerebral hemorrhage after ischemic stroke. (A-B) Representative immunoblots and quantification of PAD4 levels in the ischemic cortex 24 hours after stroke in mice treated with vehicle or tPA compared with mice undergoing sham surgery (n = 5). (C) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show Evans blue extravasation 24 hours after stroke in WT and Pad4−/− mice treated with tPA. (D) Quantification of Evans blue fluorescence intensity for each group (n = 8). (E-F) Representative immunoblots and quantification of IgG levels in capillary-depleted brain tissue at 24 hours (n = 5). (G-K) Representative immunoblots and quantification of zonula occludens-1 (ZO-1), occludin, claudin-5, and vascular endothelial–cadherin (VE-cadherin) in isolated brain microvessels (n = 5). (L) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show cerebral hemorrhage 24 hours after stroke in WT and Pad4−/− mice treated with tPA. (M) Quantification of cerebral hemorrhage by spectrophotometric hemoglobin assay (n = 8). (N-P) Effects of PAD deficiency on forelimb force test and beam walking test 24 hours after stroke (n = 10). Values are means ± standard deviation. *P < .05. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.
Figure 5.
Figure 5.
DNase I treatment or PAD4 deficiency inhibits tPA-induced upregulation of cGAS-STING and type 1 IFN signaling. (A-B) Representative immunoblots and quantification of cGAS expression in the ischemic cortex 24 hours after stroke in WT mice treated with vehicle, tPA, or tPA in combination with DNase I and in WT and Pad4−/− mice treated with tPA compared with mice undergoing sham surgery (n = 5). (C) Representative immunoblots for STING, phosphorylated (pTBK1) and total TBK1, and pIRF3 and total IRF3 expression in the ischemic cortex. (D-F) Quantification of band intensities for each group (n = 5). (G-H) Quantification of IFN-β and IL-6 levels by enzyme-linked immunosorbent assay in the ischemic cortex (n = 6). (I-J) Double immunostaining of cGAS and STING with microglial cells (Iba1), macrophages (RM0029-11H3), and neutrophils (Ly6G) in mice subjected to MCAO and tPA treatment. Scale bar, 20 μm. (K-L) Quantification of activated microglia and macrophage infiltration in the ischemic cortex (n = 6). (M-O) Quantification of the numbers of pIRF3+, IFN-β+, and IL-6+ microglial cells (Iba1) in the ischemic cortex (n = 6). Values are means ± standard deviation. *P < .05. GAPDH, glyceraldehyde-3-phosphate dehydrogenase; NS, not significant.
Figure 6.
Figure 6.
DNase I–mediated cerebrovascular protection and antihemorrhagic effects after ischemic stroke are abolished by cGAMP. (A) Representative immunoblots for STING, pTBK1 and total TBK1, and pIRF3 and total IRF3 expression in the ischemic cortex of tPA-treated mice after treatment with DNase I or DNase I in combination with cGAMP. (B-D) Quantification of band intensities for each group (n = 5). (E-F) Quantification of IFN-β and IL-6 levels by enzyme-linked immunosorbent assay in the ischemic cortex (n = 6). (G-H) Representative immunoblots and quantification of IgG levels in capillary-depleted brain tissue. (I-J) Quantification of extravascular dextran fluorescence in tPA-treated mice after treatment with DNase I or DNase I in combination with cGAMP. Scale bar, 20 μm. At 24 hours after stroke, mice were administered an intravascular injection of 40 KDa of FITC-labeled dextran, and brain sections were stained with CD31. (K) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show cerebral hemorrhage 24 hours after stroke. (L) Quantification of cerebral hemorrhage (n = 8). (M-O) Effects of DNase I or combination of DNase I and cGAMP on forelimb force test and beam walking test 24 hours after stroke in tPA-treated mice (n = 10). Values are means ± standard deviation. *P < .05. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.
Figure 7.
Figure 7.
tPA-associated BBB disruption and hemorrhage after ischemic stroke are rescued by loss of cGAS. (A) Representative immunoblots for STING, pTBK1 and total TBK1, and pIRF3 and total IRF3 expression in the ischemic cortex of WT and cGas−/− mice treated with tPA. (B-D) Quantification of band intensities for each group (n = 5). (E-F) Quantification of IFN-β and IL-6 levels by enzyme-linked immunosorbent assay in the ischemic cortex (n = 6). (G-H) Representative in vivo multiphoton microscopic images of IV injected FITC-dextran (molecular weight, 40 KDa) leakage from cortical vessels and quantification of the permeability surface (P) product of FITC-dextran at 24 hours after stroke in WT and cGas−/− mice treated with tPA (n = 6). Scale bar, 100 μm. (I-J) Representative immunoblots and quantification of IgG levels in capillary-depleted brain tissue (n = 5). (K) Representative images of the dorsal surface (upper panel) and a coronal section (bottom panel) show cerebral hemorrhage 24 hours after stroke. (L) Quantification of cerebral hemorrhage (n = 8). (M-O) Effects of cGAS deficiency on forelimb force test and beam walking test 24 hours after stroke in tPA-treated mice (n = 10). (P) Model figure showing how tPA stimulates NET formation and how NETs are sensed by microglia after ischemic stroke. Values are means ± standard deviation. *P < .05. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

References

    1. Jauch EC, Saver JL, Adams HP Jr., et al. ; Council on Clinical Cardiology . Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2013;44(3):870-947.
    1. National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group . Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333(24):1581-1587.
    1. Hacke W, Kaste M, Fieschi C, et al. ; The European Cooperative Acute Stroke Study (ECASS) . Intravenous thrombolysis with recombinant tissue plasminogen activator for acute hemispheric stroke. JAMA. 1995;274(13):1017-1025.
    1. Tanne D, Kasner SE, Demchuk AM, et al. . Markers of increased risk of intracerebral hemorrhage after intravenous recombinant tissue plasminogen activator therapy for acute ischemic stroke in clinical practice: the Multicenter rt-PA Stroke Survey. Circulation. 2002;105(14):1679-1685.
    1. Whiteley WN, Slot KB, Fernandes P, Sandercock P, Wardlaw J. Risk factors for intracranial hemorrhage in acute ischemic stroke patients treated with recombinant tissue plasminogen activator: a systematic review and meta-analysis of 55 studies. Stroke. 2012;43(11):2904-2909.
    1. Yepes M, Sandkvist M, Moore EG, Bugge TH, Strickland DK, Lawrence DA. Tissue-type plasminogen activator induces opening of the blood-brain barrier via the LDL receptor-related protein. J Clin Invest. 2003;112(10):1533-1540.
    1. Suzuki Y, Nagai N, Umemura K. A review of the mechanisms of blood-brain barrier permeability by tissue-type plasminogen activator treatment for cerebral ischemia. Front Cell Neurosci. 2016;10:2.
    1. Perez-de-Puig I, Miró-Mur F, Ferrer-Ferrer M, et al. . Neutrophil recruitment to the brain in mouse and human ischemic stroke. Acta Neuropathol. 2015;129(2):239-257.
    1. Shi K, Zou M, Jia DM, et al. . tPA mobilizes immune cells that exacerbate hemorrhagic transformation in stroke. Circ Res. 2021;128(1):62-75.
    1. Uhl B, Zuchtriegel G, Puhr-Westerheide D, et al. . Tissue plasminogen activator promotes postischemic neutrophil recruitment via its proteolytic and nonproteolytic properties. Arterioscler Thromb Vasc Biol. 2014;34(7):1495-1504.
    1. Fuchs TA, Brill A, Duerschmied D, et al. . Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. 2010;107(36):15880-15885.
    1. Savchenko AS, Borissoff JI, Martinod K, et al. . VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood. 2014;123(1):141-148.
    1. Brinkmann V, Reichard U, Goosmann C, et al. . Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532-1535.
    1. Daniel C, Leppkes M, Muñoz LE, Schley G, Schett G, Herrmann M. Extracellular DNA traps in inflammation, injury and healing. Nat Rev Nephrol. 2019;15(9):559-575.
    1. Laridan E, Denorme F, Desender L, et al. . Neutrophil extracellular traps in ischemic stroke thrombi. Ann Neurol. 2017;82(2):223-232.
    1. Kang L, Yu H, Yang X, et al. . Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat Commun. 2020;11(1):2488.
    1. Su EJ, Fredriksson L, Geyer M, et al. . Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med. 2008;14(7):731-737.
    1. Zhao BQ, Ikeda Y, Ihara H, et al. . Essential role of endogenous tissue plasminogen activator through matrix metalloproteinase 9 induction and expression on heparin-produced cerebral hemorrhage after cerebral ischemia in mice. Blood. 2004;103(7):2610-2616.
    1. Wang L, Fan W, Cai P, et al. . Recombinant ADAMTS13 reduces tissue plasminogen activator-induced hemorrhage after stroke in mice. Ann Neurol. 2013;73(2):189-198.
    1. Guo X, Shu C, Li H, et al. . Cyclic GMP-AMP ameliorates diet-induced metabolic dysregulation and regulates proinflammatory responses distinctly from STING activation. Sci Rep. 2017;7(1):6355.
    1. Suzuki Y, Nagai N, Yamakawa K, Kawakami J, Lijnen HR, Umemura K. Tissue-type plasminogen activator (t-PA) induces stromelysin-1 (MMP-3) in endothelial cells through activation of lipoprotein receptor-related protein. Blood. 2009;114(15):3352-3358.
    1. Tsuji K, Aoki T, Tejima E, et al. . Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke. 2005;36(9):1954-1959.
    1. Miller-Ocuin JL, Liang X, Boone BA, et al. . DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. OncoImmunology. 2019;8(9):e1605822.
    1. Li RHL, Ng G, Tablin F. Lipopolysaccharide-induced neutrophil extracellular trap formation in canine neutrophils is dependent on histone H3 citrullination by peptidylarginine deiminase. Vet Immunol Immunopathol. 2017;193-194:29-37.
    1. Liberale L, Bertolotto M, Minetti S, et al. . Recombinant tissue plasminogen activator (r-tPA) induces in-vitro human neutrophil migration via low density lipoprotein receptor-related protein 1 (LRP-1). Int J Mol Sci. 2020;21(19):7014.
    1. Gavillet M, Martinod K, Renella R, Wagner DD, Williams DA. A key role for Rac and Pak signaling in neutrophil extracellular traps (NETs) formation defines a new potential therapeutic target. Am J Hematol. 2018;93(2):269-276.
    1. Mackic JB, Stins M, McComb JG, et al. . Human blood-brain barrier receptors for Alzheimer’s amyloid-beta 1- 40. Asymmetrical binding, endocytosis, and transcytosis at the apical side of brain microvascular endothelial cell monolayer. J Clin Invest. 1998;102(4):734-743.
    1. Papadakis M, Hadley G, Xilouri M, et al. . Tsc1 (hamartin) confers neuroprotection against ischemia by inducing autophagy. Nat Med. 2013;19(3):351-357.
    1. Pham DL, Ban GY, Kim SH, et al. . Neutrophil autophagy and extracellular DNA traps contribute to airway inflammation in severe asthma. Clin Exp Allergy. 2017;47(1):57-70.
    1. Zhu D, Wang Y, Singh I, et al. . Protein S controls hypoxic/ischemic blood-brain barrier disruption through the TAM receptor Tyro3 and sphingosine 1-phosphate receptor. Blood. 2010;115(23):4963-4972.
    1. Xu H, Cao Y, Yang X, et al. . ADAMTS13 controls vascular remodeling by modifying VWF reactivity during stroke recovery. Blood. 2017;130(1):11-22.
    1. Cao Y, Xu H, Zhu Y, et al. . ADAMTS13 maintains cerebrovascular integrity to ameliorate Alzheimer-like pathology. PLoS Biol. 2019;17(6):e3000313.
    1. Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12(12):723-738.
    1. Li W, Chen Z, Chin I, Chen Z, Dai H. The role of VE-cadherin in blood-brain barrier integrity under central nervous system pathological conditions. Curr Neuropharmacol. 2018;16(9):1375-1384.
    1. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207(9):1853-1862.
    1. Martinod K, Demers M, Fuchs TA, et al. . Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci USA. 2013;110(21):8674-8679.
    1. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339(6121):786-791.
    1. Kerur N, Fukuda S, Banerjee D, et al. . cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat Med. 2018;24(1):50-61.
    1. Otxoa-de-Amezaga A, Miró-Mur F, Pedragosa J, et al. . Microglial cell loss after ischemic stroke favors brain neutrophil accumulation. Acta Neuropathol. 2019;137(2):321-341.
    1. Mathiesen Janiurek M, Soylu-Kucharz R, Christoffersen C, Kucharz K, Lauritzen M. Apolipoprotein M-bound sphingosine-1-phosphate regulates blood-brain barrier paracellular permeability and transcytosis. Elife. 2019;8:e49405.
    1. Zhang C, An J, Strickland DK, Yepes M. The low-density lipoprotein receptor-related protein 1 mediates tissue-type plasminogen activator-induced microglial activation in the ischemic brain. Am J Pathol. 2009;174(2):586-594.
    1. Won S, Lee JK, Stein DG. Recombinant tissue plasminogen activator promotes, and progesterone attenuates, microglia/macrophage M1 polarization and recruitment of microglia after MCAO stroke in rats. Brain Behav Immun. 2015;49:267-279.
    1. Prabhakaran S, Ruff I, Bernstein RA. Acute stroke intervention: a systematic review. JAMA. 2015;313(14):1451-1462.
    1. Guo Z, Yu S, Chen X, et al. . Suppression of NLRP3 attenuates hemorrhagic transformation after delayed rtPA treatment in thromboembolic stroke rats: Involvement of neutrophil recruitment. Brain Res Bull. 2018;137:229-240.
    1. Gautier S, Ouk T, Tagzirt M, et al. . Impact of the neutrophil response to granulocyte colony-stimulating factor on the risk of hemorrhage when used in combination with tissue plasminogen activator during the acute phase of experimental stroke. J Neuroinflammation. 2014;11:96.
    1. Gautier S, Ouk T, Petrault O, Caron J, Bordet R. Neutrophils contribute to intracerebral haemorrhages after treatment with recombinant tissue plasminogen activator following cerebral ischaemia. Br J Pharmacol. 2009;156(4):673-679.
    1. Cedervall J, Zhang Y, Huang H, et al. . Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res. 2015;75(13):2653-2662.
    1. Kastrup A, Gröschel K, Ringer TM, et al. . Early disruption of the blood-brain barrier after thrombolytic therapy predicts hemorrhage in patients with acute stroke. Stroke. 2008;39(8):2385-2387.
    1. Su EJ, Cao C, Fredriksson L, et al. . Microglial-mediated PDGF-CC activation increases cerebrovascular permeability during ischemic stroke. Acta Neuropathol. 2017;134(4):585-604.

Source: PubMed

3
Abonnieren