New Insight Into Neutrophils: A Potential Therapeutic Target for Cerebral Ischemia

Ran Chen, Xu Zhang, Lijuan Gu, Hua Zhu, Yi Zhong, Yingze Ye, Xiaoxing Xiong, Zhihong Jian, Ran Chen, Xu Zhang, Lijuan Gu, Hua Zhu, Yi Zhong, Yingze Ye, Xiaoxing Xiong, Zhihong Jian

Abstract

Ischemic stroke is one of the main issues threatening human health worldwide, and it is also the main cause of permanent disability in adults. Energy consumption and hypoxia after ischemic stroke leads to the death of nerve cells, activate resident glial cells, and promote the infiltration of peripheral immune cells into the brain, resulting in various immune-mediated effects and even contradictory effects. Immune cell infiltration can mediate neuronal apoptosis and aggravate ischemic injury, but it can also promote neuronal repair, differentiation and regeneration. The central nervous system (CNS), which is one of the most important immune privileged parts of the human body, is separated from the peripheral immune system by the blood-brain barrier (BBB). Under physiological conditions, the infiltration of peripheral immune cells into the CNS is controlled by the BBB and regulated by the interaction between immune cells and vascular endothelial cells. As the immune response plays a key role in regulating the development of ischemic injury, neutrophils have been proven to be involved in many inflammatory diseases, especially acute ischemic stroke (AIS). However, neutrophils may play a dual role in the CNS. Neutrophils are the first group of immune cells to enter the brain from the periphery after ischemic stroke, and their exact role in cerebral ischemia remains to be further explored. Elucidating the characteristics of immune cells and their role in the regulation of the inflammatory response may lead to the identification of new potential therapeutic strategies. Thus, this review will specifically discuss the role of neutrophils in ischemic stroke from production to functional differentiation, emphasizing promising targeted interventions, which may promote the development of ischemic stroke treatments in the future.

Keywords: NETs; ROS; blood-brain barrier; ischemia; neuroinflammation; neutrophils; stroke.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Chen, Zhang, Gu, Zhu, Zhong, Ye, Xiong and Jian.

Figures

Figure 1
Figure 1
The inflammatory response after cerebral ischemia. Acute ischemic events lead to oxidative stress and excitotoxicity, which lead to the activation of microglia and astrocytes and then promote the secretion of cytokines, MMPs and GFAP(glial fibrillary acidic protein). These proinflammatory signaling molecules lead to upregulation of the expression of ICAM-1 and selectins on endothelial cells and promote the entry of neutrophils, macrophages, lymphocytes and other blood-derived inflammatory cells into the ischemic area. Neutrophils are the first group of leukocytes to enter the CNS. At inflammatory sites, high levels of proinflammatory cytokines such as GM-CSF and TNF-α can enhance the release of ROS. ROS can induce the production of MMP-9 through signaling pathways, which leads to tissue damage and excessive inflammatory responses, such as microvascular basement membrane damage and BBB damage, and then cause more peripheral neutrophils to enter the damaged area, aggravating the nerve injury response. The activation of neutrophils can also lead to the release of nuclear and particulate matter, which form a wide network of DNA complexes (NETs) that further aggravate neuronal damage. In addition, DAMPs, which in turn activate microglia and peripheral immune cells (neutrophils, macrophages and lymphocytes), are released by dying neurons, resulting in the production of proinflammatory factors and thus leading to further activation of neutrophils. These pathological events lead to neuronal death and further increase damage to the ischemic brain.
Figure 2
Figure 2
(A) Active NADPH oxidase complexes promote ROS production. The activated NADPH oxidase complex uses fad to transfer electrons (e-) to oxygen to form O2− from phagocytes. O2− is produced by the activation of NADPH oxidase. O2−. O2− from phagocytes. O2− is produced by the activation of NADPH oxidase. O2− produced by NADPH oxidase can react with protons to form H2O2, which in turn produces HOCl under the action of MPO. O2− from phagocytes. O2− is produced by the activation of NADPH oxidase. O2− can react with H2O2 in the presence of Fe2+ or Cu2+ to form OH- or ROS. (B) The role of neutrophil activation in host defense and the inflammatory response. As shown on the left side, the initiation of proinflammatory cytokines or microbial molecules under physiological conditions is an immune-monitoring mechanism that ultimately enhances the antibacterial activity of neutrophils. However, as shown on the right side, excessive activation of neutrophil NADPH oxidase leads to excessive production of ROS, causing tissue damage and an excessive inflammatory response. Mediated by various proinflammatory mediators, receptor signals on the vascular endothelial surface are involved in neutrophil rolling, adhesion and endothelial barrier crossing. Phagocytosis of neutrophils leads to the activation of a series of processes, leading to the release of antimicrobial peptides, proteases, MPO and O2− from phagocytes. O2− is produced by the activation of NADPH oxidase. O2−, which is produced by the activation of NADPH oxidase, from phagocytes.
Figure 3
Figure 3
In the process of atherosclerosis, platelet-derived chemokines, such as CC chemokine ligand 5, promote the activation and recruitment of neutrophils. On the lumen side, activated neutrophils secrete granule proteins, including cathepsin G, which directly or indirectly promote the recruitment of myeloid cells. ROS and proteases secreted by neutrophils in the lumen and intima of atherosclerotic plaques lead to activation and dysfunction of the extracellular matrix in the layer and bottom of endothelial cells (ECs), resulting in leukocyte infiltration and low-density lipoprotein (LDL) extravasation. In the process of atherosclerotic plaque formation, the neutrophil-derived granule proteins antimicrobial peptides and α-defensin activate macrophages and cause them adopt a proinflammatory state. Neutrophils secrete MPO, which mediates the oxidation of low-density lipoprotein (oxLDL) and promotes the formation of foam cells. NETs stimulate plasma-like dendritic cells (pDCs) to produce atherogenic interferon (IFN)-α through NLRP3, and macrophages produce IL-1β and IL-18 but not AIM2 In the late stage of atherosclerosis, neutrophils can destroy plaque stability by secreting a network of proteins including cytotoxic histone H4, which penetrates and eventually dissolves vascular smooth muscle cells (VSMCs). Neutrophil-derived metalloproteinases can also induce VSMC death by degrading ECM. The death of VSMCs and degradation of ECM lead to the thinning of fiber caps and the formation of vulnerable plaques. Neutrophils can trigger epithelial cell desquamation, which is specifically manifested by the stimulation of endothelial cell stress and apoptosis by neutrophils, leading to endothelial cell detachment. This process is regulated by Toll-like receptor 2 (TLR2) signaling and other neural network signals in endothelial cells.
Figure 4
Figure 4
(A) The classic pathway of NETosis. Upon stimulation, ROS produced by NADPH oxidase activate the binding of PAD4 to citrullinated histones, leading to the unfolding of chromatin, and induce the translocation of enzymes such as NE and MPO in azurophilic granules to the nucleus, where they exacerbate the dissociation of histones and DNA. Finally, after the nuclear membrane and cytomembrane are dissolved, cell components including DNA, histones and granzymes are released into the intercellular space. (B) The effect of NETosis in ischemic stroke. Upon stimulation, neutrophils in the thrombus release NETs, which act as scaffolds to consolidate the thrombus. Some circulating neutrophils migrate from the blood stream into brain tissue, and they also release NETs to exacerbate neural damage.
Figure 5
Figure 5
The mechanism by which neutrophils repair the vascular endothelium. First, activated neutrophils deposit the antimicrobial peptide cathelicidin, which promotes the adhesion of circulating endothelial progenitor cells through FPR2, at the site of arterial injury. Endothelial progenitor cells recruited in this way can directly cover the injury site but also release angiogenic growth factor in a paracrine manner, thus promoting reendothelialization. Cathelicidin stimulates endothelial progenitor cells to secrete VEGF and EGF, which can promote the repair of damaged endothelial cells. Neutrophils penetrate the vascular endothelium by reacting with endothelial adhesion molecules to reduce blood flow velocity in the vasculature, and then neutrophils cross the vascular wall with the help of a series of adhesion molecules, such as P-, E-, and L-selectin; ICAM-1; and integrins (CD11b, a, and c).

References

    1. Iadecola C, Anrather J. The Immunology of Stroke: From Mechanisms to Translation. Nat Med (2011) 17:796–808. 10.1038/nm.2399
    1. Jickling GC, Liu D, Ander BP, Stamova B, Zhan X, Sharp FR. Targeting Neutrophils in Ischemic Stroke: Translational Insights From Experimental Studies. J Cereb Blood Flow Metab (2015) 35:888–901. 10.1038/jcbfm.2015.45
    1. Meisel C, Schwab JM, Prass K, Meisel A, Dirnagl U. Central Nervous System Injury-Induced Immune Deficiency Syndrome. Nat Rev Neurosci (2005) 6:775–86.
    1. Urra X, Cervera A, Villamor N, Planas AM, Chamorro A. Harms and Benefits of Lymphocyte Subpopulations in Patients With Acute Stroke. Neuroscience (2009) 158:1174–83. 10.1016/j.neuroscience.2008.06.014
    1. Kang L, Yu H, Yang X, Zhu Y, Bai X, Wang R, et al. . Neutrophil Extracellular Traps Released by Neutrophils Impair Revascularization and Vascular Remodeling After Stroke. Nat Commun (2020) 11:2488. 10.1038/s41467-020-16191-y
    1. Prince LR, Whyte MK, Sabroe I, Parker LC. The Role of TLRs in Neutrophil Activation. Curr Opin Pharmacol (2011) 11:397–403. 10.1016/j.coph.2011.06.007
    1. El-Benna J, Hurtado-Nedelec M, Marzaioli V, Marie J-C, Gougerot-Pocidalo M-A, Dang PM-C. Priming of the Neutrophil Respiratory Burst: Role in Host Defense and Inflammation. Immunol Rev (2016) 273:180–93. 10.1111/imr.12447
    1. Love S. Oxidative Stress in Brain Ischemia. Brain Pathol (1999) 9:119–31.
    1. Lenglet S, Montecucco F, Mach F. Role of Matrix Metalloproteinases in Animal Models of Ischemic Stroke. Curr Vasc Pharmacol (2015) 13:161–6.
    1. Heo SC, Kwon YW, Jang IH, Jeong GO, Lee TW, Yoon JW, et al. . Formyl Peptide Receptor 2 Is Involved in Cardiac Repair After Myocardial Infarction Through Mobilization of Circulating Angiogenic Cells. Stem Cells (2017) 35:654–65. 10.1002/stem.2535
    1. Heo SC, Kwon YW, Jang IH, Jeong GO, Yoon JW, Kim CD, et al. . WKYMVm-Induced Activation of Formyl Peptide Receptor 2 Stimulates Ischemic Neovasculogenesis by Promoting Homing of Endothelial Colony-Forming Cells. Stem Cells (2014) 32:779–90. 10.1002/stem.1578
    1. Ruhnau J, Schulze J, Dressel A, Vogelgesang A. Thrombosis, Neuroinflammation, and Poststroke Infection: The Multifaceted Role of Neutrophils in Stroke. J Immunol Res (2017) 2017:5140679. 10.1155/2017/5140679
    1. Wang Q, Tang XN, Yenari MA. The Inflammatory Response in Stroke. J Neuroimmunol (2007) 184:53–68.
    1. Yang Y, Rosenberg GA. Blood-Brain Barrier Breakdown in Acute and Chronic Cerebrovascular Disease. Stroke (2011) 42:3323–8. 10.1161/STROKEAHA.110.608257
    1. Gelderblom M, Leypoldt F, Steinbach K, Behrens D, Choe C-U, Siler DA, et al. . Temporal and Spatial Dynamics of Cerebral Immune Cell Accumulation in Stroke. Stroke (2009) 40:1849–57. 10.1161/STROKEAHA.108.534503
    1. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil Extracellular Traps License Macrophages for Cytokine Production in Atherosclerosis. Science (2015) 349:316–20. 10.1126/science.aaa8064
    1. Enzmann G, Mysiorek C, Gorina R, Cheng Y-J, Ghavampour S, Hannocks M-J, et al. . The Neurovascular Unit as a Selective Barrier to Polymorphonuclear Granulocyte (PMN) Infiltration Into the Brain After Ischemic Injury. Acta Neuropathol (2013) 125:395–412. 10.1007/s00401-012-1076-3
    1. Knowland D, Arac A, Sekiguchi KJ, Hsu M, Lutz SE, Perrino J, et al. . Stepwise Recruitment of Transcellular and Paracellular Pathways Underlies Blood-Brain Barrier Breakdown in Stroke. Neuron (2014) 82:603–17. 10.1016/j.neuron.2014.03.003
    1. Neumann J, Sauerzweig S, Rönicke R, Gunzer F, Dinkel K, Ullrich O, et al. . Microglia Cells Protect Neurons by Direct Engulfment of Invading Neutrophil Granulocytes: A New Mechanism of CNS Immune Privilege. J Neurosci (2008) 28:5965–75. 10.1523/JNEUROSCI.0060-08.2008
    1. Kim JY, Park J, Chang JY, Kim S-H, Lee JE. Inflammation After Ischemic Stroke: The Role of Leukocytes and Glial Cells. Exp Neurobiol (2016) 25:241–51.
    1. Schilling M, Besselmann M, Müller M, Strecker JK, Ringelstein EB, Kiefer R. Predominant Phagocytic Activity of Resident Microglia Over Hematogenous Macrophages Following Transient Focal Cerebral Ischemia: An Investigation Using Green Fluorescent Protein Transgenic Bone Marrow Chimeric Mice. Exp Neurol (2005) 196:290–7.
    1. Neumann J, Riek-Burchardt M, Herz J, Doeppner TR, König R, Hütten H, et al. . Very-Late-Antigen-4 (VLA-4)-Mediated Brain Invasion by Neutrophils Leads to Interactions With Microglia, Increased Ischemic Injury and Impaired Behavior in Experimental Stroke. Acta Neuropathol (2015) 129:259–77. 10.1007/s00401-014-1355-2
    1. Shi L, Sun Z, Su W, Xu F, Xie D, Zhang Q, et al. . Treg Cell-Derived Osteopontin Promotes Microglia-Mediated White Matter Repair After Ischemic Stroke. Immunity (2021). 10.1016/j.immuni.2021.04.022
    1. Schneider-Hohendorf T, Rossaint J, Mohan H, Böning D, Breuer J, Kuhlmann T, et al. . VLA-4 Blockade Promotes Differential Routes Into Human CNS Involving PSGL-1 Rolling of T Cells and MCAM-Adhesion of TH17 Cells. J Exp Med (2014) 211:1833–46. 10.1084/jem.20140540
    1. Appelgren D, Enocsson H, Skogman BH, Nordberg M, Perander L, Nyman D, et al. . Neutrophil Extracellular Traps (NETs) in the Cerebrospinal Fluid Samples From Children and Adults With Central Nervous System Infections. Cells (2019) 9. 10.3390/cells9010043
    1. Döring Y, Soehnlein O, Weber C. Neutrophil Extracellular Traps in Atherosclerosis and Atherothrombosis. Circ Res (2017) 120:736–43. 10.1161/CIRCRESAHA.116.309692
    1. Baek AE, Sutton NR, Petrovic-Djergovic D, Liao H, Ray JJ, Park J, et al. . Ischemic Cerebroprotection Conferred by Myeloid Lineage-Restricted or Global CD39 Transgene Expression. Circulation (2017) 135:2389–402. 10.1161/CIRCULATIONAHA.116.023301
    1. Chen GY, Nuñez G. Sterile Inflammation: Sensing and Reacting to Damage. Nat Rev Immunol (2010) 10:826–37. 10.1038/nri2873
    1. Chu HX, Kim HA, Lee S, Moore JP, Chan CT, Vinh A, et al. . Immune Cell Infiltration in Malignant Middle Cerebral Artery Infarction: Comparison With Transient Cerebral Ischemia. J Cereb Blood Flow Metab (2014) 34:450–9. 10.1038/jcbfm.2013.217
    1. Perez-de-Puig I, Miró-Mur F, Ferrer-Ferrer M, Gelpi E, Pedragosa J, Justicia C, et al. . Neutrophil Recruitment to the Brain in Mouse and Human Ischemic Stroke. Acta Neuropathol (2015) 129:239–57. 10.1007/s00401-014-1381-0
    1. Moxon-Emre I, Schlichter LC. Neutrophil Depletion Reduces Blood-Brain Barrier Breakdown, Axon Injury, and Inflammation After Intracerebral Hemorrhage. J Neuropathol Exp Neurol (2011) 70:218–35. 10.1097/NEN.0b013e31820d94a5
    1. Ritzel RM, Patel AR, Grenier JM, Crapser J, Verma R, Jellison ER, et al. . Functional Differences Between Microglia and Monocytes After Ischemic Stroke. J Neuroinflamm (2015) 12:106. 10.1186/s12974-015-0329-1
    1. Owen CA, Hu Z, Lopez-Otin C, Shapiro SD. Membrane-Bound Matrix Metalloproteinase-8 on Activated Polymorphonuclear Cells Is a Potent, Tissue Inhibitor of Metalloproteinase-Resistant Collagenase and Serpinase. J Immunol (2004) 172:7791–803.
    1. Rosell A, Cuadrado E, Ortega-Aznar A, Hernández-Guillamon M, Lo EH, Montaner J. MMP-9-Positive Neutrophil Infiltration Is Associated to Blood-Brain Barrier Breakdown and Basal Lamina Type IV Collagen Degradation During Hemorrhagic Transformation After Human Ischemic Stroke. Stroke (2008) 39:1121–6. 10.1161/STROKEAHA.107.500868
    1. Rosenberg GA, Estrada EY, Dencoff JE. Matrix Metalloproteinases and TIMPs Are Associated With Blood-Brain Barrier Opening After Reperfusion in Rat Brain. Stroke (1998) 29:2189–95.
    1. Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, et al. . The Involvement and Therapy Target of Immune Cells After Ischemic Stroke. Front Immunol (2019) 10:2167. 10.3389/fimmu.2019.02167
    1. Otxoa-de-Amezaga A, Gallizioli M, Pedragosa J, Justicia C, Miró-Mur F, Salas-Perdomo A, et al. . Location of Neutrophils in Different Compartments of the Damaged Mouse Brain After Severe Ischemia/Reperfusion. Stroke (2019) 50:1548–57. 10.1161/STROKEAHA.118.023837
    1. Yilmaz G, Granger DN. Leukocyte Recruitment and Ischemic Brain Injury. Neuromolecular Med (2010) 12:193–204. 10.1007/s12017-009-8074-1
    1. Faustino JV, Wang X, Johnson CE, Klibanov A, Derugin N, Wendland MF, et al. . Microglial Cells Contribute to Endogenous Brain Defenses After Acute Neonatal Focal Stroke. J Neurosci (2011) 31:12992–3001. 10.1523/JNEUROSCI.2102-11.2011
    1. Cuartero MI, Ballesteros I, Moraga A, Nombela F, Vivancos J, Hamilton JA, et al. . N2 Neutrophils, Novel Players in Brain Inflammation After Stroke: Modulation by the Pparγ Agonist Rosiglitazone. Stroke (2013) 44:3498–508. 10.1161/STROKEAHA.113.002470
    1. Mishalian I, Bayuh R, Eruslanov E, Michaeli J, Levy L, Zolotarov L, et al. . Neutrophils Recruit Regulatory T-Cells Into Tumors Via Secretion of CCL17–A New Mechanism of Impaired Antitumor Immunity. Int J Cancer (2014) 135:1178–86. 10.1002/ijc.28770
    1. Kerfoot SM, Raharjo E, Ho M, Kaur J, Serirom S, McCafferty DM, et al. . Exclusive Neutrophil Recruitment With Oncostatin M in a Human System. Am J Pathol (2001) 159:1531–9.
    1. García-Culebras A, Durán-Laforet V, Peña-Martínez C, Moraga A, Ballesteros I, Cuartero MI, et al. . Role of TLR4 (Toll-Like Receptor 4) in N1/N2 Neutrophil Programming After Stroke. Stroke (2019) 50:2922–32. 10.1161/STROKEAHA.119.025085
    1. Gorina R, Font-Nieves M, Márquez-Kisinousky L, Santalucia T, Planas AM. Astrocyte TLR4 Activation Induces a Proinflammatory Environment Through the Interplay Between MyD88-Dependent Nfκb Signaling, MAPK, and Jak1/Stat1 Pathways. Glia (2011) 59:242–55. 10.1002/glia.21094
    1. Kuang X, Wang L-F, Yu L, Li Y-J, Wang Y-N, He Q, et al. . Ligustilide Ameliorates Neuroinflammation and Brain Injury in Focal Cerebral Ischemia/Reperfusion Rats: Involvement of Inhibition of TLR4/peroxiredoxin 6 Signaling. Free Radic Biol Med (2014) 71:165–75. 10.1016/j.freeradbiomed.2014.03.028
    1. Zhang Z, Qin P, Deng Y, Ma Z, Guo H, Guo H, et al. . The Novel Estrogenic Receptor GPR30 Alleviates Ischemic Injury by Inhibiting TLR4-Mediated Microglial Inflammation. J Neuroinflamm (2018) 15:206. 10.1186/s12974-018-1246-x
    1. Jin R, Yang G, Li G. Inflammatory Mechanisms in Ischemic Stroke: Role of Inflammatory Cells. J Leukoc Biol (2010) 87:779–89. 10.1189/jlb.1109766
    1. Denorme F, Manne BK, Portier I, Eustes AS, Kosaka Y, Kile BT, et al. . Platelet Necrosis Mediates Ischemic Stroke Outcome in Mice. Blood (2020) 135:429–40. 10.1182/blood.2019002124
    1. Tajalli-Nezhad S, Karimian M, Beyer C, Atlasi MA, Azami Tameh A. The Regulatory Role of Toll-Like Receptors After Ischemic Stroke: Neurosteroids as TLR Modulators With the Focus on TLR2/4. Cell Mol Life Sci (2019) 76:523–37. 10.1007/s00018-018-2953-2
    1. Rossaint J, Herter JM, Van Aken H, Napirei M, Döring Y, Weber C, et al. . Synchronized Integrin Engagement and Chemokine Activation Is Crucial in Neutrophil Extracellular Trap-Mediated Sterile Inflammation. Blood (2014) 123:2573–84. 10.1182/blood-2013-07-516484
    1. Sionov RV, Fridlender ZG, Granot Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron (2015) 8:125–58. 10.1007/s12307-014-0147-5
    1. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. . Polarization of Tumor-Associated Neutrophil Phenotype by TGF-Beta: "N1" Versus "N2" TAN. Cancer Cell (2009) 16:183–94. 10.1016/j.ccr.2009.06.017
    1. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of Immune Suppression by Interleukin-10 and Transforming Growth Factor-Beta: The Role of T Regulatory Cells. Immunology (2006) 117:433–42.
    1. Winkler IG, Barbier V, Wadley R, Zannettino ACW, Williams S, Lévesque J-P. Positioning of Bone Marrow Hematopoietic and Stromal Cells Relative to Blood Flow In Vivo: Serially Reconstituting Hematopoietic Stem Cells Reside in Distinct Nonperfused Niches. Blood (2010) 116:375–85. 10.1182/blood-2009-07-233437
    1. Courties G, Herisson F, Sager HB, Heidt T, Ye Y, Wei Y, et al. . Ischemic Stroke Activates Hematopoietic Bone Marrow Stem Cells. Circ Res (2015) 116:407–17. 10.1161/CIRCRESAHA.116.305207
    1. Denes A, McColl BW, Leow-Dyke SF, Chapman KZ, Humphreys NE, Grencis RK, et al. . Experimental Stroke-Induced Changes in the Bone Marrow Reveal Complex Regulation of Leukocyte Responses. J Cereb Blood Flow Metab (2011) 31:1036–50. 10.1038/jcbfm.2010.198
    1. Eash KJ, Greenbaum AM, Gopalan PK, Link DC. CXCR2 and CXCR4 Antagonistically Regulate Neutrophil Trafficking From Murine Bone Marrow. J Clin Invest (2010) 120:2423–31. 10.1172/JCI41649
    1. Weisenburger-Lile D, Dong Y, Yger M, Weisenburger G, Polara GF, Chaigneau T, et al. . Harmful Neutrophil Subsets in Patients With Ischemic Stroke: Association With Disease Severity. Neurol Neuroimmunol Neuroinflamm (2019) 6:e571. 10.1212/NXI.0000000000000571
    1. Kostulas N, Kivisäkk P, Huang Y, Matusevicius D, Kostulas V, Link H. Ischemic Stroke Is Associated With a Systemic Increase of Blood Mononuclear Cells Expressing Interleukin-8 mRNA. Stroke (1998) 29:462–6.
    1. Qian H, Georges-Labouesse E, Nyström A, Domogatskaya A, Tryggvason K, Jacobsen SEW, et al. . Distinct Roles of Integrins Alpha6 and Alpha4 in Homing of Fetal Liver Hematopoietic Stem and Progenitor Cells. Blood (2007) 110:2399–407.
    1. Qian H, Tryggvason K, Jacobsen SE, Ekblom M. Contribution of Alpha6 Integrins to Hematopoietic Stem and Progenitor Cell Homing to Bone Marrow and Collaboration With Alpha4 Integrins. Blood (2006) 107:3503–10.
    1. Lapidot T, Kollet O. The Essential Roles of the Chemokine SDF-1 and Its Receptor CXCR4 in Human Stem Cell Homing and Repopulation of Transplanted Immune-Deficient NOD/SCID and NOD/SCID/B2m(null) Mice. Leukemia (2002) 16:1992–2003.
    1. Eash KJ, Means JM, White DW, Link DC. CXCR4 Is a Key Regulator of Neutrophil Release From the Bone Marrow Under Basal and Stress Granulopoiesis Conditions. Blood (2009) 113:4711–9. 10.1182/blood-2008-09-177287
    1. Christopher MJ, Liu F, Hilton MJ, Long F, Link DC. Suppression of CXCL12 Production by Bone Marrow Osteoblasts Is a Common and Critical Pathway for Cytokine-Induced Mobilization. Blood (2009) 114:1331–9. 10.1182/blood-2008-10-184754
    1. Lieschke GJ, Grail D, Hodgson G, Metcalf D, Stanley E, Cheers C, et al. . Mice Lacking Granulocyte Colony-Stimulating Factor Have Chronic Neutropenia, Granulocyte and Macrophage Progenitor Cell Deficiency, and Impaired Neutrophil Mobilization. Blood (1994) 84:1737–46.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the Site of Inflammation: The Leukocyte Adhesion Cascade Updated. Nat Rev Immunol (2007) 7:678–89.
    1. Ley K, Smith E, Stark MA. IL-17A-Producing Neutrophil-Regulatory Tn Lymphocytes. Immunol Res (2006) 34:229–42.
    1. Schwarzenberger P, Huang W, Ye P, Oliver P, Manuel M, Zhang Z, et al. . Requirement of Endogenous Stem Cell Factor and Granulocyte-Colony-Stimulating Factor for IL-17-Mediated Granulopoiesis. J Immunol (2000) 164:4783–9.
    1. Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of Apoptotic Neutrophils Regulates Granulopoiesis via IL-23 and IL-17. Immunity (2005) 22:285–94.
    1. Tyrkalska SD, Pérez-Oliva AB, Rodríguez-Ruiz L, Martínez-Morcillo FJ, Alcaraz-Pérez F, Martínez-Navarro FJ, et al. . Inflammasome Regulates Hematopoiesis Through Cleavage of the Master Erythroid Transcription Factor Gata1. Immunity (2019) 51. 10.1016/j.immuni.2019.05.005
    1. Mitroulis I, Ruppova K, Wang B, Chen L-S, Grzybek M, Grinenko T, et al. . Modulation of Myelopoiesis Progenitors Is an Integral Component of Trained Immunity. Cell (2018) 172. 10.1016/j.cell.2017.11.034
    1. Christ A, Günther P, Lauterbach MAR, Duewell P, Biswas D, Pelka K, et al. . Western Diet Triggers NLRP3-Dependent Innate Immune Reprogramming. Cell (2018) 172. 10.1016/j.cell.2017.12.013
    1. Westerterp M, Fotakis P, Ouimet M, Bochem AE, Zhang H, Molusky MM, et al. . Cholesterol Efflux Pathways Suppress Inflammasome Activation, NETosis, and Atherogenesis. Circulation (2018) 138:898–912. 10.1161/CIRCULATIONAHA.117.032636
    1. Maestrini I, Strbian D, Gautier S, Haapaniemi E, Moulin S, Sairanen T, et al. . Higher Neutrophil Counts Before Thrombolysis for Cerebral Ischemia Predict Worse Outcomes. Neurology (2015) 85:1408–16. 10.1212/WNL.0000000000002029
    1. Hernandez PA, Gorlin RJ, Lukens JN, Taniuchi S, Bohinjec J, Francois F, et al. . Mutations in the Chemokine Receptor Gene CXCR4 Are Associated With WHIM Syndrome, a Combined Immunodeficiency Disease. Nat Genet (2003) 34:70–4.
    1. Evrard M, Kwok IWH, Chong SZ, Teng KWW, Becht E, Chen J, et al. . Developmental Analysis of Bone Marrow Neutrophils Reveals Populations Specialized in Expansion, Trafficking, and Effector Functions. Immunity (2018) 48. 10.1016/j.immuni.2018.02.002
    1. Tchernychev B, Ren Y, Sachdev P, Janz JM, Haggis L, O'Shea A, et al. . Discovery of a CXCR4 Agonist Pepducin That Mobilizes Bone Marrow Hematopoietic Cells. Proc Natl Acad Sci USA (2010) 107:22255–9. 10.1073/pnas.1009633108
    1. Dabrowska S, Andrzejewska A, Lukomska B, Janowski M. Neuroinflammation as a Target for Treatment of Stroke Using Mesenchymal Stem Cells and Extracellular Vesicles. J Neuroinflamm (2019) 16:178. 10.1186/s12974-019-1571-8
    1. Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial Cells: Role of the Immune Response in Ischemic Stroke. Front Immunol (2020) 11:294. 10.3389/fimmu.2020.00294
    1. Kanazawa M, Ninomiya I, Hatakeyama M, Takahashi T, Shimohata T. Microglia and Monocytes/Macrophages Polarization Reveal Novel Therapeutic Mechanism Against Stroke. Int J Mol Sci (2017) 18. 10.3390/ijms18102135
    1. Chen A-Q, Fang Z, Chen X-L, Yang S, Zhou Y-F, Mao L, et al. . Microglia-Derived TNF-α Mediates Endothelial Necroptosis Aggravating Blood Brain-Barrier Disruption After Ischemic Stroke. Cell Death Dis (2019) 10:487. 10.1038/s41419-019-1716-9
    1. Amantea D, Micieli G, Tassorelli C, Cuartero MI, Ballesteros I, Certo M, et al. . Rational Modulation of the Innate Immune System for Neuroprotection in Ischemic Stroke. Front Neurosci (2015) 9:147. 10.3389/fnins.2015.00147
    1. Anrather J, Iadecola C. Inflammation and Stroke: An Overview. Neurotherapeutics (2016) 13:661–70. 10.1007/s13311-016-0483-x
    1. Huang Y, Chen S, Luo Y, Han Z. Crosstalk Between Inflammation and the BBB in Stroke. Curr Neuropharmacol (2020) 18:1227–36. 10.2174/1570159X18666200620230321
    1. Stowe AM, Adair-Kirk TL, Gonzales ER, Perez RS, Shah AR, Park TS, et al. . Neutrophil Elastase and Neurovascular Injury Following Focal Stroke and Reperfusion. Neurobiol Dis (2009) 35:82–90. 10.1016/j.nbd.2009.04.006
    1. del Zoppo GJ, Mabuchi T. Cerebral Microvessel Responses to Focal Ischemia. J Cereb Blood Flow Metab (2003) 23: 879–894.
    1. Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-Mediated Vascular Barrier Injury: Role of Neutrophil Extracellular Traps. Microcirculation (2017) 24. 10.1111/micc.12352
    1. Chang JJ, Emanuel BA, Mack WJ, Tsivgoulis G, Alexandrov AV. Matrix Metalloproteinase-9: Dual Role and Temporal Profile in Intracerebral Hemorrhage. J Stroke Cerebrovasc Dis (2014) 23:2498–505. 10.1016/j.jstrokecerebrovasdis.2014.07.005
    1. Yang Y, Rosenberg GA. MMP-Mediated Disruption of Claudin-5 in the Blood-Brain Barrier of Rat Brain After Cerebral Ischemia. Methods Mol Biol (2011) 762:333–45. 10.1007/978-1-61779-185-7_24
    1. Wu M-Y, Yiang G-T, Liao W-T, Tsai AP-Y, Cheng Y-L, Cheng P-W, et al. . Current Mechanistic Concepts in Ischemia and Reperfusion Injury. Cell Physiol Biochem (2018) 46:1650–67. 10.1159/000489241
    1. Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA. Matrix Metalloproteinase-Mediated Disruption of Tight Junction Proteins in Cerebral Vessels Is Reversed by Synthetic Matrix Metalloproteinase Inhibitor in Focal Ischemia in Rat. J Cereb Blood Flow Metab (2007) 27:697–709.
    1. Ikegame Y, Yamashita K, Hayashi S-i, Yoshimura S-i, Nakashima S, Iwama T. Neutrophil Elastase Inhibitor Prevents Ischemic Brain Damage via Reduction of Vasogenic Edema. Hypertens Res (2010) 33:703–7. 10.1038/hr.2010.58
    1. Yang Y, Thompson JF, Taheri S, Salayandia VM, McAvoy TA, Hill JW, et al. . Early Inhibition of MMP Activity in Ischemic Rat Brain Promotes Expression of Tight Junction Proteins and Angiogenesis During Recovery. J Cereb Blood Flow Metab (2013) 33:1104–14. 10.1038/jcbfm.2013.56
    1. Nauseef WM. How Human Neutrophils Kill and Degrade Microbes: An Integrated View. Immunol Rev (2007) 219.
    1. Hampton MB, Kettle AJ, Winterbourn CC. Inside the Neutrophil Phagosome: Oxidants, Myeloperoxidase, and Bacterial Killing. Blood (1998) 92:3007–17.
    1. Babior BM. Phagocytes and Oxidative Stress. Am J Med (2000) 109:33–44.
    1. Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR, et al. . Infliximab and Methotrexate in the Treatment of Rheumatoid Arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis With Concomitant Therapy Study Group. N Engl J Med (2000) 343:1594–602.
    1. Minozzi S, Bonovas S, Lytras T, Pecoraro V, González-Lorenzo M, Bastiampillai AJ, et al. . Risk of Infections Using Anti-TNF Agents in Rheumatoid Arthritis, Psoriatic Arthritis, and Ankylosing Spondylitis: A Systematic Review and Meta-Analysis. Expert Opin Drug Saf (2016) 15:11–34.
    1. Bafutto M, Oliveira EC, Rezende Filho J. Use of Vitamin D With Anti-Tumor Necrosis Factor Therapy for Crohn's Disease. Gastroenterology Res (2020) 13:101–6. 10.14740/gr1264
    1. Ding Z, Liu S, Wang X, Deng X, Fan Y, Sun C, et al. . Hemodynamic Shear Stress via ROS Modulates PCSK9 Expression in Human Vascular Endothelial and Smooth Muscle Cells and Along the Mouse Aorta. Antioxid Redox Signal (2015) 22:760–71. 10.1089/ars.2014.6054
    1. Zhu L, He P. fMLP-Stimulated Release of Reactive Oxygen Species From Adherent Leukocytes Increases Microvessel Permeability. Am J Physiol Heart Circ Physiol (2006) 290:H365–72.
    1. Jacobi J, Sela S, Cohen HI, Chezar J, Kristal B. Priming of Polymorphonuclear Leukocytes: A Culprit in the Initiation of Endothelial Cell Injury. Am J Physiol Heart Circ Physiol (2006) 290:H2051–8.
    1. Lauterbach M, O'Donnell P, Asano K, Mayadas TN. Role of TNF Priming and Adhesion Molecules in Neutrophil Recruitment to Intravascular Immune Complexes. J Leukoc Biol (2008) 83:1423–30. 10.1189/jlb.0607421
    1. Guice KS, Oldham KT, Caty MG, Johnson KJ, Ward PA. Neutrophil-Dependent, Oxygen-Radical Mediated Lung Injury Associated With Acute Pancreatitis. Ann Surg (1989) 210:740–7.
    1. Conner WC, Gallagher CM, Miner TJ, Tavaf-Motamen H, Wolcott KM, Shea-Donohue T. Neutrophil Priming State Predicts Capillary Leak After Gut Ischemia in Rats. J Surg Res (1999) 84:24–30.
    1. Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: Friend and Foe for Ischemic Stroke. J Neuroinflamm (2019) 16:142. 10.1186/s12974-019-1516-2
    1. Global, Regional, and National Age-Sex-Specific Mortality for 282 Causes of Death in 195 Countries and Territories, 1980-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet (2018) 392:1736–88. 10.1016/S0140-6736(18)32203-7
    1. Xiao J, Chen L, Melander O, Orho-Melander M, Nilsson J, Borné Y, et al. . Circulating Vimentin Is Associated With Future Incidence of Stroke in a Population-Based Cohort Study. Stroke (2021) 52:937–44. 10.1161/STROKEAHA.120.032111
    1. Amin-Hanjani S, See AP, Du X, Rose-Finnell L, Pandey DK, Chen Y-F, et al. . Natural History of Hemodynamics in Vertebrobasilar Disease: Temporal Changes in the VERiTAS Study Cohort. Stroke (2020) 51:3295–301. 10.1161/STROKEAHA.120.029909
    1. Chistiakov DA, Kashirskikh DA, Khotina VA, Grechko AV, Orekhov AN. Immune-Inflammatory Responses in Atherosclerosis: The Role of Myeloid Cells. J Clin Med (2019) 8. 10.3390/jcm8111798
    1. Soehnlein O, Lindbom L, Weber C. Mechanisms Underlying Neutrophil-Mediated Monocyte Recruitment. Blood (2009) 114:4613–23. 10.1182/blood-2009-06-221630
    1. Alard J-E, Ortega-Gomez A, Wichapong K, Bongiovanni D, Horckmans M, Megens RTA, et al. . Recruitment of Classical Monocytes Can Be Inhibited by Disturbing Heteromers of Neutrophil HNP1 and Platelet CCL5. Sci Transl Med (2015) 7:317ra196. 10.1126/scitranslmed.aad5330
    1. Rasmuson J, Kenne E, Wahlgren M, Soehnlein O, Lindbom L. Heparinoid Sevuparin Inhibits -Induced Vascular Leak Through Neutralizing Neutrophil-Derived Proteins. FASEB J (2019) 33:10443–52. 10.1096/fj.201900627R
    1. Delporte C, Boudjeltia KZ, Noyon C, Furtmüller PG, Nuyens V, Slomianny M-C, et al. . Impact of Myeloperoxidase-LDL Interactions on Enzyme Activity and Subsequent Posttranslational Oxidative Modifications of apoB-100. J Lipid Res (2014) 55:747–57. 10.1194/jlr.M047449
    1. Quillard T, Araújo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and Neutrophils Potentiate Endothelial Stress, Apoptosis and Detachment: Implications for Superficial Erosion. Eur Heart J (2015) 36:1394–404. 10.1093/eurheartj/ehv044
    1. Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, et al. . Flow Perturbation Mediates Neutrophil Recruitment and Potentiates Endothelial Injury via TLR2 in Mice: Implications for Superficial Erosion. Circ Res (2017) 121:31–42. 10.1161/CIRCRESAHA.117.310694
    1. Papayannopoulos V. Neutrophil Extracellular Traps in Immunity and Disease. Nat Rev Immunol (2018) 18:134–47. 10.1038/nri.2017.105
    1. Yang Q-W, Lu F-L, Zhou Y, Wang L, Zhong Q, Lin S, et al. . HMBG1 Mediates Ischemia-Reperfusion Injury by TRIF-Adaptor Independent Toll-Like Receptor 4 Signaling. J Cereb Blood Flow Metab (2011) 31:593–605. 10.1038/jcbfm.2010.129
    1. Kim S-W, Lee H, Lee H-K, Kim I-D, Lee J-K. Neutrophil Extracellular Trap Induced by HMGB1 Exacerbates Damages in the Ischemic Brain. Acta Neuropathol Commun (2019) 7:94. 10.1186/s40478-019-0747-x
    1. Martinez NE, Zimmermann TJ, Goosmann C, Alexander T, Hedberg C, Ziegler S, et al. . Tetrahydroisoquinolines: New Inhibitors of Neutrophil Extracellular Trap (NET) Formation. Chembiochem (2017) 18:888–93. 10.1002/cbic.201600650
    1. Jorch SK, Kubes P. An Emerging Role for Neutrophil Extracellular Traps in Noninfectious Disease. Nat Med (2017) 23:279–87. 10.1038/nm.4294
    1. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, et al. . Histone Hypercitrullination Mediates Chromatin Decondensation and Neutrophil Extracellular Trap Formation. J Cell Biol (2009) 184:205–13. 10.1083/jcb.200806072
    1. Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, et al. . Neutrophil Extracellular Traps Contain Calprotectin, a Cytosolic Protein Complex Involved in Host Defense Against Candida Albicans. PloS Pathog (2009) 5:e1000639. 10.1371/journal.ppat.1000639
    1. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, et al. . Extracellular DNA Traps Promote Thrombosis. Proc Natl Acad Sci U.S.A. (2010) 107:15880–5. 10.1073/pnas.1005743107
    1. Folco EJ, Mawson TL, Vromman A, Bernardes-Souza B, Franck G, Persson O, et al. . Neutrophil Extracellular Traps Induce Endothelial Cell Activation and Tissue Factor Production Through Interleukin-1α and Cathepsin G. Arterioscler Thromb Vasc Biol (2018) 38:1901–12. 10.1161/ATVBAHA.118.311150
    1. Franck G, Mawson TL, Folco EJ, Molinaro R, Ruvkun V, Engelbertsen D, et al. . Roles of PAD4 and NETosis in Experimental Atherosclerosis and Arterial Injury: Implications for Superficial Erosion. Circ Res (2018) 123:33–42. 10.1161/CIRCRESAHA.117.312494
    1. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. . Neutrophil Extracellular Traps Kill Bacteria. Science (2004) 303:1532–5.
    1. Dwivedi N, Radic M. Citrullination of Autoantigens Implicates NETosis in the Induction of Autoimmunity. Ann Rheum Dis (2014) 73:483–91. 10.1136/annrheumdis-2013-203844
    1. Ebrahimi F, Giaglis S, Hahn S, Blum CA, Baumgartner C, Kutz A, et al. . Markers of Neutrophil Extracellular Traps Predict Adverse Outcome in Community-Acquired Pneumonia: Secondary Analysis of a Randomised Controlled Trial. Eur Respir J (2018) 51. 10.1183/13993003.01389-2017
    1. Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A, et al. . Coronary Neutrophil Extracellular Trap Burden and Deoxyribonuclease Activity in ST-Elevation Acute Coronary Syndrome Are Predictors of ST-Segment Resolution and Infarct Size. Circ Res (2015) 116:1182–92. 10.1161/CIRCRESAHA.116.304944
    1. Schauer C, Janko C, Munoz LE, Zhao Y, Kienhöfer D, Frey B, et al. . Aggregated Neutrophil Extracellular Traps Limit Inflammation by Degrading Cytokines and Chemokines. Nat Med (2014) 20:511–7. 10.1038/nm.3547
    1. Barber GN. STING-Dependent Cytosolic DNA Sensing Pathways. Trends Immunol (2014) 35:88–93. 10.1016/j.it.2013.10.010
    1. Kato K, Omura H, Ishitani R, Nureki O. Cyclic GMP-AMP as an Endogenous Second Messenger in Innate Immune Signaling by Cytosolic DNA. Annu Rev Biochem (2017) 86:541–66. 10.1146/annurev-biochem-061516-044813
    1. Knight JS, Luo W, O'Dell AA, Yalavarthi S, Zhao W, Subramanian V, et al. . Peptidylarginine Deiminase Inhibition Reduces Vascular Damage and Modulates Innate Immune Responses in Murine Models of Atherosclerosis. Circ Res (2014) 114:947–56. 10.1161/CIRCRESAHA.114.303312
    1. Nakagomi N, Nakagomi T, Kubo S, Nakano-Doi A, Saino O, Takata M, et al. . Endothelial Cells Support Survival, Proliferation, and Neuronal Differentiation of Transplanted Adult Ischemia-Induced Neural Stem/Progenitor Cells After Cerebral Infarction. Stem Cells (2009) 27:2185–95. 10.1002/stem.161
    1. Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, et al. . Endothelial Cells Stimulate Self-Renewal and Expand Neurogenesis of Neural Stem Cells. Science (2004) 304:1338–40.
    1. Brea D, Sobrino T, Ramos-Cabrer P, Castillo J. [Reorganisation of the Cerebral Vasculature Following Ischaemia]. Rev Neurol (2009) 49:645–54.
    1. Hermann DM, Zechariah A. Implications of Vascular Endothelial Growth Factor for Postischemic Neurovascular Remodeling. J Cereb Blood Flow Metab (2009) 29:1620–43. 10.1038/jcbfm.2009.100
    1. Rundhaug JE. Matrix Metalloproteinases and Angiogenesis. J Cell Mol Med (2005) 9:267–85.
    1. Dumont CM, Piselli JM, Kazi N, Bowman E, Li G, Linhardt RJ, et al. . Factors Released From Endothelial Cells Exposed to Flow Impact Adhesion, Proliferation, and Fate Choice in the Adult Neural Stem Cell Lineage. Stem Cells Dev (2017) 26:1199–213. 10.1089/scd.2016.0350
    1. Greenberg DA, Jin K. From Angiogenesis to Neuropathology. Nature (2005) 438:954–9.
    1. Zhao B-Q, Wang S, Kim H-Y, Storrie H, Rosen BR, Mooney DJ, et al. . Role of Matrix Metalloproteinases in Delayed Cortical Responses After Stroke. Nat Med (2006) 12:441–5.
    1. Zhu W, Khachi S, Hao Q, Shen F, Young WL, Yang G-Y, et al. . Upregulation of EMMPRIN After Permanent Focal Cerebral Ischemia. Neurochem Int (2008) 52:1086–91. 10.1016/j.neuint.2007.11.005
    1. Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, Wu Z, et al. . Apolipoprotein E Controls Cerebrovascular Integrity via Cyclophilin a. Nature (2012) 485:512–6. 10.1038/nature11087
    1. Herz J, Hagen SI, Bergmüller E, Sabellek P, Göthert JR, Buer J, et al. . Exacerbation of Ischemic Brain Injury in Hypercholesterolemic Mice Is Associated With Pronounced Changes in Peripheral and Cerebral Immune Responses. Neurobiol Dis (2014) 62:456–68. 10.1016/j.nbd.2013.10.022
    1. Qian S, You S, Sun Y, Wu Q, Wang X, Tang W, et al. . Remnant Cholesterol and Common Carotid Artery Intima-Media Thickness in Patients With Ischemic Stroke. Circ Cardiovasc Imaging (2021) 14:e010953. 10.1161/CIRCIMAGING.120.010953
    1. Herz J, Sabellek P, Lane TE, Gunzer M, Hermann DM, Doeppner TR. Role of Neutrophils in Exacerbation of Brain Injury After Focal Cerebral Ischemia in Hyperlipidemic Mice. Stroke (2015) 46:2916–25. 10.1161/STROKEAHA.115.010620
    1. Hristov M, Zernecke A, Bidzhekov K, Liehn EA, Shagdarsuren E, Ludwig A, et al. . Importance of CXC Chemokine Receptor 2 in the Homing of Human Peripheral Blood Endothelial Progenitor Cells to Sites of Arterial Injury. Circ Res (2007) 100:590–7.
    1. Kim M-H, Gorouhi F, Ramirez S, Granick JL, Byrne BA, Soulika AM, et al. . Catecholamine Stress Alters Neutrophil Trafficking and Impairs Wound Healing by β2-Adrenergic Receptor-Mediated Upregulation of IL-6. J Invest Dermatol (2014) 134:809–17. 10.1038/jid.2013.415
    1. Luo Y, Xia L-X, Li Z-L, Pi D-F, Tan X-P, Tu Q. Early Neutrophil-to-Lymphocyte Ratio Is a Prognostic Marker in Acute Minor Stroke or Transient Ischemic Attack. Acta Neurol Belg (2020). 10.1007/s13760-020-01289-3
    1. Zhao L, Dai Q, Chen X, Li S, Shi R, Yu S, et al. . Neutrophil-To-Lymphocyte Ratio Predicts Length of Stay and Acute Hospital Cost in Patients With Acute Ischemic Stroke. J Stroke Cerebrovasc Dis (2016) 25:739–44. 10.1016/j.jstrokecerebrovasdis.2015.11.012
    1. Tao C, Wang J, Hu X, Ma J, Li H, You C. Clinical Value of Neutrophil to Lymphocyte and Platelet to Lymphocyte Ratio After Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care (2017) 26:393–401. 10.1007/s12028-016-0332-0
    1. Ortega-Gomez A, Salvermoser M, Rossaint J, Pick R, Brauner J, Lemnitzer P, et al. . Cathepsin G Controls Arterial But Not Venular Myeloid Cell Recruitment. Circulation (2016) 134:1176–88.
    1. Yang Y, Rosenberg GA. Matrix Metalloproteinases as Therapeutic Targets for Stroke. Brain Res (2015) 1623:30–8. 10.1016/j.brainres.2015.04.024
    1. Drechsler M, de Jong R, Rossaint J, Viola JR, Leoni G, Wang JM, et al. . Annexin A1 Counteracts Chemokine-Induced Arterial Myeloid Cell Recruitment. Circ Res (2015) 116:827–35. 10.1161/CIRCRESAHA.116.305825
    1. Zhang Y, Jian W, He L, Wu J. Externalized Histone H4: A Novel Target That Orchestrates Chronic Inflammation by Inducing Lytic Cell Death. Acta Biochim Biophys Sin (Shanghai) (2020) 52:336–8. 10.1093/abbs/gmz165
    1. Vajen T, Koenen RR, Werner I, Staudt M, Projahn D, Curaj A, et al. . Blocking CCL5-CXCL4 Heteromerization Preserves Heart Function After Myocardial Infarction by Attenuating Leukocyte Recruitment and NETosis. Sci Rep (2018) 8:10647. 10.1038/s41598-018-29026-0
    1. Certo M, Endo Y, Ohta K, Sakurada S, Bagetta G, Amantea D. Activation of RXR/Pparγ Underlies Neuroprotection by Bexarotene in Ischemic Stroke. Pharmacol Res (2015) 102:298–307. 10.1016/j.phrs.2015.10.009
    1. Ludewig P, Sedlacik J, Gelderblom M, Bernreuther C, Korkusuz Y, Wagener C, et al. . Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 Inhibits MMP-9-Mediated Blood-Brain-Barrier Breakdown in a Mouse Model for Ischemic Stroke. Circ Res (2013) 113:1013–22. 10.1161/CIRCRESAHA.113.301207
    1. Han Z, Li L, Wang L, Degos V, Maze M, Su H. Alpha-7 Nicotinic Acetylcholine Receptor Agonist Treatment Reduces Neuroinflammation, Oxidative Stress, and Brain Injury in Mice With Ischemic Stroke and Bone Fracture. J Neurochem (2014) 131:498–508. 10.1111/jnc.12817
    1. Neumann S, Shields NJ, Balle T, Chebib M, Clarkson AN. Innate Immunity and Inflammation Post-Stroke: An α7-Nicotinic Agonist Perspective. Int J Mol Sci (2015) 16:29029–46. 10.3390/ijms161226141
    1. Sreeramkumar V, Adrover JM, Ballesteros I, Cuartero MI, Rossaint J, Bilbao I, et al. . Neutrophils Scan for Activated Platelets to Initiate Inflammation. Science (2014) 346:1234–8. 10.1126/science.1256478
    1. Paulin N, Viola JR, Maas SL, de Jong R, Fernandes-Alnemri T, Weber C, et al. . Double-Strand DNA Sensing Aim2 Inflammasome Regulates Atherosclerotic Plaque Vulnerability. Circulation (2018) 138:321–3. 10.1161/CIRCULATIONAHA.117.033098

Source: PubMed

3
Abonnieren