key: cord-0743212-lbslkq4j authors: Zhang, Qingguang; Ling, Shang; Hu, Kaili; Liu, Jun; Xu, Jin-Wen title: Role of the renin-angiotensin system in NETosis in the coronavirus disease 2019 (COVID-19) date: 2022-02-14 journal: Biomed Pharmacother DOI: 10.1016/j.biopha.2022.112718 sha: 19837fb5f1b7408b43507ec7d8bdab04321d8184 doc_id: 743212 cord_uid: lbslkq4j Myocardial infarction and stroke are the leading causes of death in the world. Numerous evidence has confirmed that hypertension promotes thrombosis and induces myocardial infarction and stroke. Recent findings reveal that neutrophil extracellular traps (NETs) are involved in the induction of myocardial infarction and stroke. Meanwhile, patients with severe COVID-19 suffer from complications such as myocardial infarction and stroke with pathological signs of NETs. Due to the extremely low amount of virus detected in the blood and remote organs (e.g., heart, brain and kidney) in a few cases, it is difficult to explain the mechanism by which the virus triggers NETosis, and there may be a different mechanism than in the lung. A large number of studies have found that the renin-angiotensin system regulates the NETosis at multiple levels in patients with COVID-19, such as endocytosis of SARS-COV-2, abnormal angiotensin II levels, neutrophil activation and procoagulant function at multiple levels, which may contribute to the formation of reticular structure and thrombosis. The treatment of angiotensin-converting enzyme inhibitors (ACEI), angiotensin II type 1 receptor blockers (ARBs) and neutrophil recruitment and active antagonists helps to regulate blood pressure and reduce the risk of net and thrombosis. The review will explore the possible role of the angiotensin system in the formation of NETs in severe COVID-19. Severe COVID-19 patients develop acute respiratory distress syndrome, which is a life-threatening situation. Neutrophil extracellular traps (NETs) are considered to be the core factor in the pathophysiology and clinical manifestations of thrombosis and inflammatory damage to the lung, heart, and brain tissues. Studies have revealed that SARS-CoV-2, a coronavirus, directly triggers the formation of NETs in patients with pulmonary embolism [1] [2] [3] [4] [5] . The process is dependent on angiotensin-converting enzyme 2 (ACE2), serine protease, viral replication and protein arginine deiminase 4 (PAD4) [1] . The levels of free DNA, myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (Cit-H3) in sera from patients with COVID-19 are elevated [5] . All autopsy specimens showed pulmonary infiltration of neutrophils and the Abbreviations: AAA, abdominal aortic aneurysm; ACE2, angiotensin-converting enzyme 2; ACEI, ACE inhibitors; ADAM 17, a disintegrin and metalloprotease domain 17; AGT, angiotensinogen; Ang 1-7, angiotensin 1-7; Ang II, angiotensin II; ARBs, angiotensin receptor blockers; AREs, AU-rich elements; AT1R, angiotensin II type 1 receptor; CDK4/6, cyclin-dependent kinases 4/6; Cit-H3, citrullinated histone H3; cPLA2, cytosolic phospholipase A2; dsDNA, double-stranded DNA; G-CSF, granulocyte colony stimulating factor; GSDMD, gasdermin D; HOCl, hypochlorous acid; IL-8, interleukin-8; LDGs, low-density granulocytes; LDNs, low-density neutrophils; MLKL, mixed lineage kinase domain-like; MPO-DNA, myeloperoxidase-DNA complex; NADPH oxidase, nicotinamide adenine dinucleotide phosphateoxidase; NE, neutrophil elastase; NETosis, a program for formation of neutrophil extracellular traps; NETs, neutrophil extracellular traps; NEP, neprilysin; NF-κB, Nuclear factor kappaB; PAD4, protein arginine deiminase 4; PDE4, phosphodiesterase 4; PKC, protein kinase C; POP/ PEP, prolyl oligopeptidase/ prolyl endopeptidase; RIPK, receptor interacting protein kinase; PRCP, prolyl carboxypeptidase; RGD motif, Arg-Gly-Asp motif; TNF-α, tumor necrosis factor alpha. presence of NETs [1] [2] [3] [4] . The low-density neutrophils (LDNs) that cause NETosis have been identified as the driver of lung injury, especially microvascular thrombosis and respiratory dysfunction [4] . Severe SARS-CoV-2 infection also causes microvascular thrombosis in the lung, heart, brain, pancreas, and other tissues, and induces multiorgan dysfunction, such as ST-elevation myocardial infarction [6] [7] [8] , stroke [9, 10] , and new-onset diabetes [11, 12] . Alveolar type II epithelial cells are the main target cells after SARS-CoV-2 infection. Cell death and marked innate immune responses during infection may lead to alveolar damage [13, 14] , alveolar collapse [15] , restricted oxygen diffusion exchange [16] and silent hypoxia [16] . SARS-CoV-2 infection also causes excessive infiltration and activation of neutrophils leading to the formation of NETs, and induces local thrombosis leading to tissue damage. In severe cases, NET components are detected in the circulation and thereby promote microthrombosis and NET-associated sequelae in remote organs [17] [18] [19] . SARS-CoV-2 uses ACE2 as a receptor to enter the cell and disrupts the balance of the angiotensin system, resulting in angiotensin II (Ang II) mediating tissue microvascular damage [20, 21] . The accumulated clinical data shows that ACE inhibitors (ACEI) or angiotensin receptor blockers (ARBs) are beneficial for the treatment of COVID-19 [22, 23] . However, there is a gap in understanding between the disorder of the angiotensin system and the formation of NETs triggered by SARS-CoV-2. This review will focus on the angiotensin system and discuss its coordination effect on SARS-CoV-2 on the priming and triggering of NETs. Neutrophils are the first cellular responders to invading pathogens and provide early immune protection. Neutrophils enter the circulation from the bone marrow and are eliminated by specialized macrophages from the circulation due to cell aging. The process exhibits different cell phenotypes and functional changes. Under inflammatory conditions, the life cycle of neutrophils will be prolonged [24] . According to the development of neutrophils, they are divided into immature neutrophils, mature neutrophils and hypersegmented neutrophils. Immature neutrophils have no distinctly divided serrated nuclei, while hypersegmented neutrophils are aged and degenerated neutrophils containing 6-10 segmental nuclei. They also can be divided into low-density granulocytes (LDGs) and normal-density granulocytes by ficoll gradient centrifugation. LDGs usually contain immature neutrophils and mature neutrophils, displaying characteristics of pro-inflammatory and immunosuppressive, and showing huge cell phenotypic heterogeneity, high oxidative burst, and functional plasticity in inflammatory conditions [25] . For the phenotypic characteristics of neutrophils in COVID-19 patients, multiple studies have confirmed the heterogeneity of neutrophils in their peripheral blood and the expansion of myeloid cells with a decrease in the proportion of basophils and eosinophils [26] [27] [28] . Even in the recovery period after 3 months of infection, the level of LDNs in the blood remained elevated [29] . The LDNs in COVID-19 patients express intermediate levels of CD16, show pro-inflammatory characteristics, spontaneously form NETs, and enhance the ability of phagocytosis and cytokine production [30] . The use of high-dimensional flow cytometry and Hierarchical clustering based on marker expression revealed a CD16 bright population in patients with moderate COVID-19, showing higher expression levels of CD11b, CD177, and CD66b. Conversely, the immature CD16 dim population enriched in severe COVID-19 patients shows a higher heterogeneous phenotype, such as CD66b and CD11b [26] . The conclusion of the trend of expanding to LDNs is also confirmed with another high-dimensional flow cytometry study, reflecting the characteristics of emergency myelopoiesis, neutrophil recruitment and activation [27] . Furthermore, a transcriptome analysis of single cells in whole blood has provided important information that in the severe COVID-19 patient group, neutrophil activation-related features are significantly enriched, and granulocytes show increased inflammation and inhibitory features at the same time. When comparing the severe and mild samples from the 1st to 10th days after the onset of symptoms, differential expression analysis has also identified 314-upregulated genes and 703-downregulated genes. For example, in the granulocytes of patients with severe COVID-19, the expressions of immature neutrophil-related markers CD15, NETs formation-related enzyme PAD4, pro-inflammatory MMP8, S100A8/9, and NLRC4 are elevated [31] . Similarly, another whole blood single-cell transcriptome study has also identified gene signatures that are highly correlated with neutrophils [32] . Together, these studies have demonstrated that both neutrophil characteristics of phenotypic and gene expression tend to be heterogeneous and activated. In patients with severe COVID-19, NETosis triggered by SARS-CoV-2 was ACE2-dependent [1] . It is reported that numerous viruses have been detected in neutrophils and even activate neutrophils to produce NETs [33] . As a single-stranded RNA virus, SARS-CoV-2 is quickly recognized by endosomal toll-like receptors (TLR) 7/8 after entering the cell, which activates the TLR-MyD88 signaling pathway, oxidative burst, NETs formation, and pro-inflammatory cytokines release [19, [34] [35] [36] (Fig. 1 ). Multiple reports of IgA vasculitis have been reported following SARS-CoV-2 infections [37, 38] . IgA immune complexes can be recognized by FcαRI, a member of the Fc receptor immunoglobulin superfamily, and activate NETs in rheumatoid diseases [39, 40] . The immune complex composed of SARS-CoV-2-IgA is used as one of the pathways to initiate NETs [41] . LDNs display a mature primed heterogeneous phenotype, and the function of β2 integrin can partly explain the heterogeneity of neutrophils. αMβ2 integrins (CD11b/CD18, complement receptor 3, or Mac-1) is an important marker of low-density neutrophils and has the ability to initiate NETs formation. There are pieces of evidence that low-density neutrophils display high expression of CD11b in both healthy and diseased individuals [42, 43] . αMβ2 integrins (CD11b/CD18) has a sequence that senses the RGD motif (Arg-Gly-Asp) [44] [45] [46] . Interestingly, the spike protein of SARS-CoV-2 contains an RGD motif in its receptor binding domain, which other coronaviruses so far [47, 48] . The RGD motif provides a basis for SARS-CoV-2 to invade host cells by binding to integrins. The high expression of integrins in the lung and all other important organs finally leads to systemic reactions after SARS-CoV-2 infections. αMβ2 (CD11b/CD18) activates neutrophils and regulates the plasticity and fate of neutrophils through "outside-in" signaling [49] (Fig. 1) . Multiple studies have shown that αMβ2 integrins (CD11b/CD18) mediates the release of NETs induced by hantavirus, Aspergillus fumigatus, and immune complexes [48] [49] [50] [51] [52] , which also provides indirect evidence that SARS-CoV-2 can induce the NET formation through αMβ2 integrins "outside-in" signaling. Integrins combine with their ligands to induce cell adhesion and produce "outside-in" signaling, which participates in slow rolling, enhanced adhesion, transendothelial migration and signal transduction, promotes oxidative burst, cytokine production, proliferation, survival, differentiation, degranulation, and cell polarization [53] [54] [55] . Compared with monocytes, neutrophils highly express α9β1 integrins, which upregulates neutrophil activation and promotes NETosis, thrombosis, and inflammation [56, 57] . In neutrophils, β1 integrin activation promotes β2 integrin-mediated adhesion, and β2 integrin engagement induces the surface expression of β1 integrin, which identified an interaction and regulation relationship between β2 and β1 integrins [58, 59] . αVβ3 integrins also contributes to the speed and linear movement of neutrophils [60] , and regulates the oxidative burst in the human neutrophils adhered to fibrinogen [61] . Because of the RGD motif in the viral spike protein, cell surface integrin may act as a co-receptor for SARS-CoV-2. Studies have revealed that the cytoplasmic tail of β3 integrin mediates the endocytosis and transport of SARS-CoV-2 by binding to ACE2 in cell-free systems [62, 63] . Numerous studies have confirmed a cytokine storm in COVID-19, with TNF-α, IL-1β, IL-6, IL-8, and MCP-1 rising dramatically in the lungs and circulation [17, 18] . Excessive Ang-II leads to multiple deleterious cardiovascular consequences, such as increased blood pressure, cardiomyocyte apoptosis, macrophage infiltration, and secretion of proinflammatory cytokines TNF-α, IL-1β, IL-6, and MCP-1 [64] [65] [66] [67] [68] . Activated renin-angiotensin also induces a prothrombotic state resulting from an imbalance of coagulation and fibrinolysis [69, 70] . Elevated Ang II level is strongly associated with SARS-CoV-2 viral load, inflammation and lung damage in severe COVID-19 patients [71, 72] . A meta-analysis showed that ACEI/ARB therapy does not increase the risk of SARS-CoV-2 infection, but instead it is associated with a reduced risk of severe COVID-19 and mortality [73] . A review pointed out that although SARS-CoV-2 genomic material was not detected in the blood of most patients with COVID-19, very low levels of the virus were found in a few cases [74] . SARS-CoV-2 RNA has also been detected in tissues and cells such as liver, kidney, and heart [74] . Another analysis of SARS-CoV-2 infection in 4103-donors of blood also showed that RNAemia was rare in plasma (0.66%) [75] . Moreover, NETosis can be triggered by the plasma of patients with COVID-19 and inhibited by the SYK inhibitor fostamatinib/R406 [76] , suggesting the existence of a possible non-SARS-CoV-2-triggered mechanism of NETosis. Therefore, clarifying the mechanisms of the renin-angiotensin system in the pathogenesis of COVID-19 is beneficial for improving cardiovascular comorbidities. ACE2, which acts as a receptor for SARS-CoV-2, is highly expressed in the heart, blood vessels, and kidneys. ACE2 converts Ang II to angiotensin 1-7 (Ang 1-7) (Fig. 2) . The catalytically active extracellular domain of ACE2 falls off, presenting in a soluble form in the circulation. However, there are still many factors that can affect the plasma levels of soluble ACE2 (sACE2). Several studies have revealed that patients with heart failure, lower left ventricular ejection fraction and systolic dysfunction, atrial fibrillation, aortic stenosis, obstructive coronary artery disease and cardiac remodeling have higher ACE2 plasma activity [77] [78] [79] [80] . The plasma sACE2 levels are positively correlated with an increased risk of heart failure, myocardial infarction, stroke, and diabetes [81] [82] [83] . Severe COVID-19 patients are often accompanied by cardiovascular disease, while the high ACE2 level of cardiovascular disease patients further raises the incidence rate of severe COVID-19. Recent clinical investigations of critically ill patients with COVID-19 showed significantly heightened levels of plasma ACE2 [84] [85] [86] [87] [88] , which is associated with an increase in the maximum severity of disease within 28 days in admitted COVID-19 patients. Similarly, the plasma ACE2 of hypertensive patients with COVID-19 is dramatically increased compared with non-hypertension patients [84] . In severe COVID-19 patients, the plasma ACE2 levels presented a parabolic increase over time with a peak value of 9-11 days after hospitalization [89] . These findings suggest that the increase in plasma sACE2 is caused by endocytosis and cleavage of ACE2 ectodomain after binding to SARS-CoV-2, so that ACE2 expression on the cell membrane is then downregulated. At the circulating cell level, this imbalance expression of ACE2 membrane protein mainly comes from the monocytes [90] . The binding of spike glycoprotein of SARS-CoV-2 to ACE2 is highly similar to that of SARS-CoV [91, 92] . In vivo experimental SARS-CoV infection in wild-type mice resulted in a significant decrease in lung ACE2 expression [93] . SARS-CoV-2 infection leads to various symptoms including asymptomatic severe hypoxemia, which is also known as silent hypoxia [94, 95] , which promotes the expression of hypoxia-induced factor 1α (HIF-1α) [96] , thereby inhibiting ACE2 expression [97, 98] through a pathway that activates miRNA let-7b and LncRNA ALT1 [99, 100] (Fig. 3) . Furthermore, up-regulated HIF-1α expression stimulates ADAM17 expression [101, 102] , a cleavage enzyme in the extracellular region of ACE2, which leads to an increase in circulating ACE2 (Fig. 3) . Although individual studies have shown that the circulating Ang II concentration of COVID-19 patients is lower or unchanged than that of healthy people [103, 104] , most studies have demonstrated a considerable increase in circulating Ang II levels in severe COVID-19 patients. The serum concentration of Ang II was positively correlated with the severity of COVID-19 [88, 105, 106] . These results are derived from the induction of endocytosis due to the binding of SARS-CoV-2 to the ACE2 receptor, and the increased shedding of the extracellular domain protein of ACE2, which slows down the degradation of Ang II [107, 108] (Fig. 3) . It is also attributed to the fact that hypoxia promotes the ACE expression in endothelial cells [109, 110] , which induces Ang II production. However, the clinical findings of Ang 1-7 blood concentration in COVID-19 patients are chaotic. This discrepancy mainly showed that the concentration decreased [105, 111] , remained unchanged [91, 104] , and elevated [86, 89, 112] . In the angiotensin system, another participant inhibited by HIF-1α is prolyl oligopeptidase (POP) /neprilysin on the endothelial surface [113] , which converts angiotensin I to Ang 1-7 [114] [115] [116] (Fig. 3) . These contradictory results on circulating Ang 1-7 levels are difficult to explain from the inhibition of ACE2 and POP by HIF-1α. Moreover, the expression or activity of the third enzyme that forms Ang 1-7, prolyl carboxypeptidase (PRCP), has not been reported to be regulated by hypoxia or SARS-CoV-2. Capillary endothelial cells occupy a very high proportion in the lung, heart and brain, where are the organs in which NETosis is prevalent in COVID-19 patients. Earlier studies have determined that the capillary endothelial cells of the lung make up 30% of lung cells.Furthermore, interspecies comparisons of cellular characteristics in the alveolar region of normal human, baboon, and rat lungs showed that the proportion, average thickness, size, and surface area of alveolar cells were relatively constant [117] . In the heart, 60% of cardiac non-cardiomyocytes are endothelial cells [118] . A more detailed analysis revealed that endothelial cells accounted for 12.2% of the atrial tissue and 7.8% of the ventricular tissue [119] . The blood-brain barrier is a unique structure dominated by capillary endothelial cells that distribute throughout the brain. The proportion of endothelial cells is 25% in the cerebral cortex and 12.2% in the spinal cord [120] . Obviously, capillary endothelial cells occupy a very high proportion in the lung, heart and brain. A study found that the lungs of Covid-19 patients exhibited a unique vascular signature of severe endothelial damage associated with intracellular virus [121] , which causes capillary flow obstruction and restricts the diffusive exchange of oxygen in the lungs and other tissues causing silent hypoxia. The patients with COVID-19 had extensive thrombosis with microvascular disease, and the incidence of alveolar-capillary micro-thrombosis was 9 folds higher than that of patients with influenza, even if they both caused acute respiratory distress syndrome [121] , suggesting that capillary occlusion marked by NETs is a characteristic of COVID-19. ACE is an enzyme that catalyzes the production of Ang II and is expressed on the surface of the endothelial cells and certain cells [122] . It has been reported that the monoclonal antibody (mAb) 9B9 using ACE selectively targets the endothelium in human lung tissue with high density after systemic injection [123, 124] . ACE staining in the heart was confined to endothelial cells and distributed gradients along the vascular tree around the entire arterial endothelial ring, but not in veins [125] . Using in situ hybridization detection, ACE mRNA was shown to be expressed in the choroid plexus, caudate putamen and cerebellum [126] . In the subfornix organ (SFO), the expression of ACE is found in ependymal cells, vascular endothelial cells, some glial cells, and neurons on the lumen surface [127] . However, in contrast to the cytological specificity of ACE expression, ACE2 expression is present in almost all organs and cells [128] . Therefore, the endothelial cell regional arrangements in tissues and the cell-type-specific expression of ACE are the biological basis of local Ang II high expression and high secretion after SARS-CoV-2 infection, and also the reason for high NETosis incidence in the lung, heart, and brain. Silent hypoxia induced by SARS-CoV-2 infection induces rapid local high expression of ACE, which in turn promotes the production of Ang II [96, 129] . Numerous early studies have demonstrated that hypoxia promotes the plasma activities of Ang II and renin [130] [131] [132] , which is consistent with the elevated plasma Ang II levels in COVID-19 patients. Long-term chronic hypoxia, such as sleep apnea syndrome and high-altitude hypoxia, leads to hypertension [133, 134] , heart failure [135, 136] , and stroke [137] . Elevated plasma Ang II also induced NETosis [138] . Previous studies have suggested that the oxidative burst activity of neutrophils in patients with essential hypertension is significantly increased [139, 140] . Recently, investigates have reported that patients with hypertension display heterogeneity of neutrophils, and the phenotypic activation of CD11b, CD66b, and CD63 of LDNs, which have significantly higher respiratory bursts than that of healthy individuals [141] . The up-regulation of CD63 in LDNs is consistent with the release of azurophilic granules [142] , indicating that these cells have been activated and degranulated [143] . In addition, increased PD-L1 expression is a feature of LDGs. AngII increases PD-L1 mRNA stability through the binding of HuR to the AU-rich elements (AREs) in the 3'-UTR, thereby increasing the expression of PD-L1 mRNA and protein, and the percentage of CD11b + Ly6G + granulocytes in mice [144] (Fig. 4) . In terms of initiating neutrophil heterogeneity, ACE binds β1 integrin in an RGD independent manner to trigger outside-in signaling [145, 146] , while Ang II promotes the expression of β1 integrin [147, 148] . Multiple studies have demonstrated that Ang II enhances the expression of αvβ3 integrins in neutrophils via the NF-κB signaling pathway [149, 150] . In a study of spontaneously hypertensive rats, the angiotensin II type 1 receptor (AT1R) antagonists improved Mac-1 (CD11b/CD18) expression of neutrophils and increased cerebral microvascular permeability [151] . Notably, the expression of CD11b in neutrophils does not increase at rest in patients with essential hypertension. However, after PMA stimulation, the neutrophils expressed CD11b faster and the primary granulosa accelerated the speed of exocytosis [152] . Hypertension and Ang II have the characteristics of inducing a low-grade chronic inflammation, which may play a priming effect on the formation of NETs as same as pro-inflammatory cytokines, such as IL-1α, IL-6, IL-8, TNF-α, G-CSF, and MIF [153] [154] [155] [156] [157] (Fig. 4) . As is well known that malignant hypertension is the main cause of myocardial infarction, and ischemic stroke [158] [159] [160] [161] . Naturally, as a blood pressure regulator, Ang II is the initiator of myocardial infarction Fig. 4 . In remote organs, angiotensin II initiates NETosis. Angiotensin II activates NETosis by enhancing the heterogeneity of neutrophils, eliciting cell sensitivity, and triggering signal transduction, respectively. Ang II: angiotensin II. and ischemic stroke. In the process of inducing myocardial infarction and ischemic stroke, Ang II not only promotes the production of oxidative stress [162, 163] , endoplasmic reticulum stress [162] , and inflammation [164, 165] , but also regulates necroptosis [166] . Based on these observations, ACE2 and Ang (1− 7) antagonize the actions of the renin-angiotensin-system, and thereby improve acute pulmonary embolism [167, 168] , inhibit inflammation [169] , suppress oxidative stress [170] , and eliminate apoptosis caused by endoplasmic reticulum stress [171] . Accumulated data have demonstrated that NETs are also found in myocardial infarction [172, 173] , ischemic stroke [174, 175] , and pulmonary embolism [176, 177] . Early studies have documented that AT1Rs are present on neutrophils [151, 178] . Neutrophils produce oxidative burst when stimulated by Ang II [179, 180] . In acute myocardial infarction, Ang II stimulates rapid neutrophil recruitment and infiltration by releasing CXC chemokines, such as IL-8 [181] . Past results indicate that Ang II has the potential to induce NETosis. Immunofluorescence staining showed that cit-H3 of plasma NET biomarker, myeloperoxidase (MPO) and neutrophil elastase (NE) expression increased significantly in a mouse model of abdominal aortic aneurysm (AAA) induced by Ang II infusion. Simultaneously, the circulating levels of double-stranded DNA (dsDNA) are elevated [182] . In human patients of AAA, cit-H3 in the blood and aortic tissue increased significantly compared with that of healthy controls. During the repair phase after aneurysm surgery, the blood cit-H3 of patients decreased markedly [183] . The use of PAD4 inhibitors, YW3-56 or GSK484, resulted in NETosis inhibition and further growth of aneurysms induced by Ang II [182, 183] . Furthermore, at the cellular level, treatment of neutrophils with the NADPH oxidase inhibitor DPI or the PAD4 inhibitor Cl-amidine before Ang II stimulation also significantly reduced Ang II-mediated NETase [138] (Fig. 4) . The NETosis requires multiple signal transductions, including cell pyroptosis, necroptosis, mitosis, and oxidative burst to achieve the needs for protease dissociation of Azurophilic granules, cytolysis, histone cleavage, lamin B1 degradation, chromatin depolymerization, and nuclear membrane rupture. Recent findings revealed the signal transduction and cellular events required for NETosis (Fig. 5) . (1) Caspase1/4/11 and gasdermin D (GSDMD) in pyroptosis are required by NETosis and contribute to cytolysis. Presently, three research groups indicated that the pore-forming protein GSDMD cleaved by Caspase 11 is essential for NET release [184] [185] [186] . Moreover, Ang II-induced spleen tyrosine kinase (SYK) activates inflammasome activation [187] [188] [189] . Oxidative stress induced by Ang II causes the activation of JNK in human neutrophils [190, 191] , which is necessary for NETosis [192] . In short, it can be considered that Ang II, as a pathological susceptibility factor, can drive the NLRP3-caspase 1-GSDMD pathway to initiate NETs [184] [185] [186] . (2) A large number of studies have revealed that FAS/FASL and other receptors mediated the cytosolic apoptotic complex IIb to transduce necroptosis signals [193, 194] . In necroptosis, the receptor interacting protein kinase (RIPK) 1/3 cascade phosphorylates a pseudokinase called mixed lineage kinase domain-like (MLKL), which oligomerizes on the plasma membrane, initiats the membrane rupture, and regulates the ion flow [193, 194] . MLKL also plays a key role in activating the downstream PAD4 activity and NETosis [195, 196] . Ang II triggers RIPK3-MLKL-mediated necroptosis through elevating Fas/FasL expression [197] . (3) A new report indicates that NETosis is accompanied by lamin B1 degradation and nuclear membrane rupture, which requires the participation of cyclin-dependent kinases 4/6 (CDK4/6). [198] . Although there are multiple pathways such as NFκB, mTOR, and STATs, the ERK1/2 pathway is fundamental for activating cyclin D1-CDK4/6 [199, 200] . It has been reported that ERK and p38 MAP kinases are involved in the activation of cPLA2 and NADPH oxidase, and that cPLA2 is required for Ang II to activate NADPH oxidase in neutrophils [190, 201] . Ang II directly enhances the expression of cyclin D1 and CDK4 [202, 203] . A clinical study showed elevated transcription levels of CDK4 and CDK6 in LDNs. The high ratio of LDNs in rheumatoid patients is mainly in the G2/S phase of the cell cycle [204] . Protein kinase C (PKC) α is another driving factor for the disintegration of laminin B and nuclear membrane rupture to promote NETosis [205, 206] . (4) Oxidative burst is a very critical factor for NETosis [207] . PKCα is required for full assembly of NADPH oxidase and activation of the oxidative burst in neutrophils [208] , while the process is stimulated by Ang II [179] . Rac2 is another key player in the NADPH oxidase assembly and activation of neutrophils. Deletion of Rac2 will lead to an impaired NADPH oxidase activity [209, 210] . Moreover, its GTPase activity is closely related to NETosis [211, 212] . The activation of Rac2 can also be exerted by AngII through calcineurin, mitogen activated protein kinases, and other pathways [191] . (5) In Azurophilic granules of neutrophils, MPO is activated by oxidative bursts to produce hypochlorous acid (HOCl). HOCl targets plasmalogen to generate 2-chloro fatty acids (2-ClFA). Human neutrophils treated with physiological levels of 2-ClFA initiate the NETosis process without activation and degranulation [213] . After MPO is activated, it also triggers the dissociation of neutrophil elastase (NE) from membrane-associated complexes to the cytoplasm to hydrolyze F-actin, and then transfer to the nucleus to cleavage histones [214] . Although the results of regulation of signal transduction by Ang II do not come entirely from neutrophils, an overview of signal pathways of NETosis can still be observed. A large number of clinical applications have proved that AT1R blockers and ACE inhibitors are effective and beneficial in improving severe COVID-19 symptoms, not only for serious clinical events induced by Ang II in elderly patients, such as hypertension, diabetes, or congestive heart failure [215] [216] [217] , and also for NETosis and thrombosis promoted by the imbalance of the angiotensin system [218, 219] . Experimental studies have revealed that AT1R receptor blockers hinder Ang II-induced AAA formation [220, 221] , which is one of the important comorbidities of COVID-19 [222] . NETs are important participants in the pathogenesis of AAA [182, 183] . Animal experiments also confirmed that AT1R blockers impede NETosis [218] . NETosis is a PAD4-dependent phenomenon, thus, the inhibitors of the protein citrullination catalytic enzyme PAD4 have shown the effect of intercepting NETosis in the Ang II-induced AAA [182, 183] . Phosphodiesterase 4 (PDE4) is a major cyclic-3',5'-adenosine monophosphate (cAMP) metabolizing enzyme. Its blockers roflumilast and rolipram can increase the level of intracellular cAMP and inhibit the formation of NETs [223, 224] as well as Ang II-induced AAA [225] . In the case study, 4 patients with COVID-19 were treated with the oral PDE4 inhibitor Apremilast. Preliminary evidence proves that apremilast is safe and beneficial in the treatment of SARS-CoV-2 pneumonia. It is also suitable for patients with older age, cardiovascular comorbidities, greater extent of lung involvement, and higher IL-6 levels [226] . Many reports pointed out that PKA, a cAMP-dependent protein kinase, negatively regulates NADPH oxidase activation and prevents oxidative burst [227, 228] . Unlike the NETosis triggered by SARS-CoV-2 in the lungs, the viral infection and detection rate in remote organs were extremely low, suggesting a different mechanism for the NETosis. Alveolar epithelial injury and alveolar collapse lead to severe diffusion-limited oxygen exchange and silent hypoxia, which in turn results in high ACE expression, low membrane ACE2 protein levels in endothelial-rich remote organs, and ultimately causes high local Ang II levels. Combined with the enhanced neutrophil heterogeneity of the COVID-19, NETosis is triggered by Ang II. Ang II-mediated NETs-induced thromboembolism causes severe damage to organs such as the heart and brain. The clinical efficacy and benefit of ACEIs and ARBs also partially illustrate the critical role of the renin-angiotensin system in NETosis. This review is based on our idea. The manuscript, or part of it, neither has been published, nor is currently under consideration for publication by any other journal. The submitting author should declare that the co-authors have read the manuscript and approved its submission to Biomedicine & Pharmacotherapy. This work was supported by grants from the Specialized Research Fund for the National Natural Science Foundation of China (81973511). The authors declare that they have no competing interests. The authors declare that they have no competing interests. SARS-CoV-2-triggered neutrophil extracellular traps mediate COVID-19 pathology Targeting potential drivers of COVID-19: neutrophil extracellular traps Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19 Neutrophil extracellular traps in fatal COVID-19-associated lung injury Neutrophil extracellular traps in COVID-19 Assessment of neutrophil extracellular traps in coronary thrombus of a case series of patients with COVID-19 and myocardial infarction COVID coronary vascular thrombosis: correlation with neutrophil but not endothelial activation SARS-COV-2 colonizes coronary thrombus and impairs heart microcirculation bed in asymptomatic SARS-CoV-2 positive subjects with acute myocardial infarction Clinical characteristics and outcomes of COVID-19 patients with a history of stroke in Wuhan Cerebral venous sinus thrombosis in COVID-19 Patients: a multicenter study and review of literature SARS-CoV-2 infection of the pancreas promotes thrombofibrosis and is associated with new-onset diabetes Expression of SARS-CoV-2 entry factors in the pancreas of normal organ donors and individuals with COVID-19 Respiratory epithelial cell responses to SARS-CoV-2 in COVID-19 Markers of endothelial and epithelial pulmonary injury in mechanically ventilated COVID-19 ICU patients Ultra-high-resolution computed tomography can demonstrate alveolar collapse in novel coronavirus (COVID-19) pneumonia SARS CoV-2 related microvascular damage and symptoms during and after COVID-19: consequences of capillary transit-time changes, tissue hypoxia and inflammation COVID-19: lung-centric immunothrombosis Lungcentric inflammation of COVID-19: potential modulation by vitamin D Severe acute respiratory syndrome coronavirus 2, COVID-19, and the renin-angiotensin system: pressing needs and best research practices COVID-19 and microvascular disease: pathophysiology of SARS-CoV-2 infection with focus on the renin-angiotensin system Targeting the renin-angiotensin signaling pathway in COVID-19: unanswered questions, opportunities, and challenges COVID-19 and renin-angiotensin system inhibition: role of angiotensin converting enzyme 2 (ACE2) -Is there any scientific evidence for controversy? Neutrophil heterogeneity: implications for homeostasis and pathogenesis The enigma of low-density granulocytes in humans: complexities in the characterization and function of LDGs during disease Highdimensional profiling reveals phenotypic heterogeneity and disease-specific alterations of granulocytes in COVID-19 Characterization of lowdensity granulocytes in COVID-19 High expression of neutrophil and monocyte CD64 with simultaneous lack of upregulation of adhesion receptors CD11b, CD162, CD15, CD65 on neutrophils in severe COVID-19 Mild and asymptomatic COVID-19 convalescents present long-term endotype of immunosuppression associated with neutrophil subsets possessing regulatory functions A specific low-density neutrophil population correlates with hypercoagulation and disease severity in hospitalized COVID-19 patients Giamarellos-Bourboulis, T. Ulas, German COVID-19 omics initiative (DeCOI), disease severity-specific neutrophil signatures in blood transcriptomes stratify COVID-19 patients Single-Cell transcriptome analysis highlights a role for neutrophils and inflammatory macrophages in the pathogenesis of severe COVID-19 The emerging role of neutrophil extracellular traps in severe acute respiratory syndrome coronavirus 2 (COVID-19) SARS-CoV-2-associated ssRNAs activate inflammation and immunity via TLR7/8 SARS-CoV-2 RBD trimer protein adjuvanted with Alum-3M-052 protects from SARS-CoV-2 infection and immune pathology in the lung Increased peripheral blood neutrophil activation phenotypes and NETosis in critically ill COVID-19 patients: a case series and review of the literature Reactivation of IgA vasculitis after COVID-19 vaccination IgA complexes in plasma and synovial fluid of patients with rheumatoid arthritis induce neutrophil extracellular traps via FcαRI Sputum neutrophil extracellular trap subsets associate with IgA anti-citrullinated protein antibodies in subjects at-risk for rheumatoid arthritis IgA potentiates NETosis in response to viral infection Induction of low density and up-regulation of CD11b expression of neutrophils and eosinophils by dextran sedimentation and centrifugation Frontline science: extracellular CIRP generates a proinflammatory Ly6G+CD11bhi subset of lowdensity neutrophils in sepsis Conservation of structural and functional domains in complement component C3 of Xenopus and mammals CR3 (CD11b/CD18) expresses one binding site for Arg-Gly-Asp-containing peptides and a second site for bacterial lipopolysaccharide Ligand specificity of purified complement receptor type three (CD11b/CD18, αmβ2, Mac-1). Indirect effects of an Arg-Gly-Asp (RGD) sequence A potential role for integrins in host cell entry by SARS-CoV-2 SARS-CoV-2 attachment to host cells is possibly mediated via RGDintegrin interaction in a calcium-dependent manner and suggests pulmonary EDTA chelation therapy as a novel treatment for COVID 19 β2 integrin regulation of neutrophil functional plasticity and fate in the resolution of inflammation Schönrich, β2 integrin mediates hantavirus-induced release of neutrophil extracellular traps Mac-1 triggers neutrophil DNA extracellular trap formation to Aspergillus fumigatus independently of PAD4 histone citrullination Immobilized immune complexes induce neutrophil extracellular trap release by human neutrophil granulocytes via FcγRIIIB and Mac-1 The ins and outs of leukocyte integrin signaling Impaired integrin-dependent function in Wiskott-Aldrich syndrome protein-deficient murine and human neutrophils Triggering of chloride ion efflux from human neutrophils as a novel function of leukocyte β2 integrins: relationship with spreading and activation of the respiratory burst Targeting myeloid-cell specific integrin α9β1 inhibits arterial thrombosis in mice Targeting myeloidspecific integrin α9β1 improves short-and long-term stroke outcomes in murine models with preexisting comorbidities by limiting thrombosis and inflammation β1 integrin activation on human neutrophils promotes β2 integrin-mediated adhesion to fibronectin Engagement of β2 integrins induces surface expression of β1 integrin receptors in human neutrophils Transendothelial migration enhances integrin-dependent human neutrophil chemokinesis αVβ3 integrin regulation of respiratory burst in fibrinogen adherent human neutrophils Cytoplasmic short linear motifs in ACE2 and integrin β3 link SARS-CoV-2 host cell receptors to mediators of endocytosis and autophagy Short linear motif candidates in the cell entry system used by SARS-CoV-2 and their potential therapeutic implications Physiological regulation of pro-inflammatory cytokines expression in rat cardiovascular tissues by sympathetic nervous system and angiotensin II Angiotensin II regulates the synthesis of proinflammatory cytokines and chemokines in the kidney Increased angiotensin II-induced hypertension and inflammatory cytokines in mice lacking angiotensin-converting enzyme N domain activity Involvement of endoplasmic reticulum stress in angiotensin II-induced NLRP3 inflammasome activation in human renal proximal tubular cells in vitro Autophagy attenuates angiotensin ii-induced pulmonary fibrosis by inhibiting redox imbalance-mediated nod-like receptor family pyrin domain containing 3 inflammasome activation Angiotensin II, tissue factor and the thrombotic paradox of hypertension Stimulation of plasminogen activator inhibitor in vivo by infusion of angiotensin II. Evidence of a potential interaction between the renin-angiotensin system and fibrinolytic function Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury COVID-19 and heart failure: from infection to inflammation and angiotensin II stimulation. Searching for evidence from a new disease Association of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers with risk of COVID-19, inflammation level, severity, and death in patients with COVID-19: a rapid systematic review and meta-analysis Evidence of SARS-CoV-2 infection in cells, tissues, and organs and the risk of transmission through transplantation Analysis of current SARS-CoV-2 infection in a large population of blood donors evidenced that RNAemia is rare in plasma Fostamatinib inhibits neutrophils extracellular traps induced by COVID-19 Patient Plasma: a potential therapeutic Soluble angiotensin-converting enzyme 2 in human heart failure: relation with myocardial function and clinical outcomes Angiotensin converting enzyme 2 activity and human atrial fibrillation: increased plasma angiotensin converting enzyme 2 activity is associated with atrial fibrillation and more advanced left atrial structural remodeling Plasma ACE2 activity predicts mortality in aortic stenosis and is associated with severe myocardial fibrosis Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease Soluble angiotensin converting enzyme 2 levels in chronic heart failure is associated with decreased exercise capacity and increased oxidative stress-mediated endothelial dysfunction Plasma ACE2 and risk of death or cardiometabolic diseases: a case-cohort analysis Clinical and proteomic correlates of plasma ACE2 (angiotensin-converting enzyme 2) in human heart failure Soluble Angiotensin-converting enzyme 2, cardiac biomarkers, structure, and function, and cardiovascular events (from the atherosclerosis risk in communities study) Plasma ACE2 predicts outcome of COVID-19 in hospitalized patients Increased blood angiotensin converting enzyme 2 activity in critically ill COVID-19 patients Soluble angiotensin converting enzyme 2 (ACE2) is upregulated and soluble endothelial nitric oxide synthase (eNOS) is downregulated in COVID-19-induced acute respiratory distress syndrome (ARDS) Soluble angiotensin-converting enzyme 2 is transiently elevated in COVID-19 and correlates with specific inflammatory and endothelial markers A dramatic rise in serum ACE2 activity in a critically ill COVID-19 patient Expression of ACE2, soluble ACE2, angiotensin I, Ang II and angiotensin-(1-7) Is modulated in COVID-19 patients Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirusinduced lung injury Improved survival following wardbased non-invasive pressure support for severe hypoxia in a cohort of frail patients with COVID-19: retrospective analysis from a UK teaching hospital Predictors and clinical outcomes of silent hypoxia in COVID-19 patients, a single-center retrospective cohort study Hypoxic and pharmacological activation of HIF inhibits SARS-CoV-2 infection of lung epithelial cells Role of HIF-1α in the regulation ACE and ACE2 expression in hypoxic human pulmonary artery smooth muscle cells MiRNA let-7b promotes the development of hypoxic pulmonary hypertension by targeting ACE2 A human long non-coding RNA ALT1 controls the cell cycle of vascular endothelial cells via ACE2 and cyclin D1 pathway Hypoxia-induced ADAM 17 expression is mediated by RSK1-dependent C/EBPβ activation in human lung fibroblasts Experimental data using candesartan and captopril indicate no double-edged sword effect in COVID-19 TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein A pilot study to assess the circulating renin-angiotensin system in COVID-19 acute respiratory failure Efficacy of serum angiotensin II levels in prognosis of patients with coronavirus disease Reninangiotensin system dysregulation in critically ill patients with acute respiratory distress syndrome due to COVID-19: a preliminary report Elevation of plasma angiotensin II level is a potential pathogenesis for the critically ill COVID-19 patients Hypertension as a sequela in patients of SARS-CoV-2 infection The effect of oxygen tension on the in vitro production and release of angiotensin-converting enzyme by bovine pulmonary artery endothelial cells Hypoxia stimulates endothelial cell angiotensin-converting enzyme antigen synthesis Coronavirus disease 2019 is associated with low circulating plasma levels of angiotensin 1 and angiotensin 1,7 Increased circulating levels of angiotensin-(1-7) in severely ill COVID-19 patients Regulation and differential expression of neutral endopeptidase 24.11 in human endothelial cells Hypoxia decreases lung neprilysin expression and increases pulmonary vascular leak Decreased neprilysin and pulmonary vascular remodeling in chronic obstructive pulmonary disease Hypoxia-induced downregulation of neprilysin by histone modification in mouse primary cortical and hippocampal neurons Cell number and cell characteristics of the normal human lung Revisiting cardiac cellular composition Cells of the adult human heart The cellular composition and glia-neuron ratio in the spinal cord of a human and a nonhuman primate: comparison with other species and brain regions Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in covid-19 Localization of angiotensin converting enzyme (kininase II). II. Immunocytochemistry and immunofluorescence Antibodymediated lung endothelium targeting: in vivo model on primates Cloning and characterization of a single-chain fragment of monoclonal antibody to ACE suitable for lung endothelial targeting Expression of cardiac angiotensin-converting enzyme after myocardial infarction Expression of angiotensin converting enzyme mRNA in rat brain Dual peroxidase and colloidal gold-labeling study of angiotensin converting enzyme and angiotensin-like immunoreactivity in the rat subfornical organ Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis Moderate intensity aerobic exercise potential favorable effect against COVID-19: the role of renin-angiotensin system and immunomodulatory effects Plasma angiotensin II levels in hypoxic and hypovolemic stress in unanesthetized rabbits Correlation of plasma angiotensin II concentration and plasma renin activity during acute hypoxia in dogs Endogenous angiotensin II in the regulation of hypoxic pulmonary vasoconstriction in anaesthetized dogs Salt-sensitive hypertension induced by decoy of transcription factor hypoxia-inducible factor-1alpha in the renal medulla Sustained systemic arterial hypertension induced by extended hypobaric hypoxia Obstructive sleep apnoea, intermittent hypoxia and heart failure with a preserved ejection fraction Oxygen, oxidative stress, hypoxia, and heart failure Obstructive sleep apnea syndrome is a risk factor for stroke Angiotensin II triggers release of neutrophil extracellular traps, linking thromboinflammation with essential hypertension Enhanced activation of the respiratory burst oxidase in neutrophils from hypertensive patients Elevated neutrophil respiratory burst activity in essential hypertensive patients Heterogeneity of neutrophils in arterial hypertension Membrane surface antigen expression on neutrophils: a reappraisal of the use of surface markers for neutrophil activation Characterization of neutrophil subsets in healthy human pregnancies Angiotensin II contributes to intratumoral immunosuppressionvia induction of PD-L1 expression in non-small cell lung carcinoma Angiotensinconverting enzyme is involved in outside-in signaling in endothelial cells Angiotensin converting enzyme (ACE) and ACE2 bind integrins and ACE2 regulates integrin signaling High glucose and angiotensin II increase β1 integrin and integrin-linked kinase synthesis in cultured mouse podocytes Protein kinase Cε mediates angiotensin IIinduced activation of β1-integrins in cardiac fibroblasts Angiotensin II and α(v) β(3) integrin expression in rat neonatal cardiac fibroblasts Angiotensin II stimulates endothelial integrin β3 expression via nuclear factor-kappaB activation Role of angiotensin II type 1 receptor in the leucocytes and endothelial cells of brain microvessels in the pathogenesis of hypertensive cerebral injury Rapid fusion of granules with neutrophil cell membranes in hypertensive patients may increase vascular damage Macrophage migration inhibitory factor (MIF) enhances hypochlorous acid production in phagocytic neutrophils Stimulation and priming of human neutrophils by granulocyte colony-stimulating factor and granulocytemacrophage colony-stimulating factor: qualitative and quantitative differences Priming and stimulation of bovine neutrophils by recombinant human interleukin-1α and tumor necrosis factor α Interleukin-8-induced priming of neutrophil oxidative burst requires sequential recruitment of NADPH oxidase components into lipid rafts Association of the renin-sodium profile with the risk of myocardial infarction in patients with hypertension Long-term prognosis after acute myocardial infarction in patients with a history of arterial hypertension TRACE study group Hypertension in acute ischemic stroke: a compensatory mechanism or an additional damaging factor? Acute post-stroke blood pressure relative to premorbid levels in intracerebral haemorrhage versus major ischaemic stroke: a populationbased study Modulation of oxidative stress by a selective inhibition of angiotensin II type 1 receptors in MI rats Oxidative stress in the infarcted heart: role of de novo angiotensin II production ER stress in the brain subfornical organ mediates angiotensin-dependent hypertension Upregulation of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) in wall of ruptured human cerebral aneurysms: preliminary results Cyclooxygenase-2 in myocardium stimulation by angiotensin-II in cultured cardiac fibroblasts and role at acute myocardial infarction Klotho attenuates angiotensin II-induced cardiotoxicity through suppression of necroptosis and oxidative stress RhoA-Rho associated kinase signaling leads to renin-angiotensin system imbalance and angiotensin converting enzyme 2 has a protective role in acute pulmonary embolism The angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptor axis: a potential target for treating thrombotic diseases Angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas axis protects against lung fibrosis by inhibiting the MAPK/NF-κB pathway The angiotensin-converting enzyme 2/angiotensin (1-7)/Mas axis protects against lung fibroblast migration and lung fibrosis by inhibiting the NOX4-derived ROS-mediated RhoA/Rho kinase pathway Abrogation of ER stress-induced apoptosis of alveolar epithelial cells by angiotensin 1-7 Neutrophils, neutrophil extracellular traps and interleukin-17 associate with the organisation of thrombi in acute myocardial infarction Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction Neutrophil extracellular traps are increased in patients with acute ischemic stroke: prognostic significance Neutrophil extracellular traps in ischemic stroke thrombi Prothrombotic fibrin clot properties associated with NETs formation characterize acute pulmonary embolism patients with higher mortality risk Neutrophil extracellular traps promote fibrous vascular occlusions in chronic thrombosis Nonpeptide antagonists of AT1 receptor for angiotensin II delay the onset of acute respiratory distress syndrome Angiotensin II-induced oxidative burst is fluvastatin sensitive in neutrophils of patients with hypercholesterolemia The association between angiotensin II-induced free radical generation and membrane fluidity in neutrophils of patients with metabolic syndrome Angiotensin II induces neutrophil accumulation in vivo through generation and release of CXC chemokines Inhibition of peptidyl arginine deiminase 4-dependent neutrophil extracellular trap formation reduces angiotensin ii-induced abdominal aortic aneurysm rupture in mice Histone citrullination as a novel biomarker and target to inhibit progression of abdominal aortic aneurysms Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps Gasdermin D plays a vital role in the generation of neutrophil extracellular traps Expression and mechanism of spleen tyrosine kinase activation by angiotensin II and its implication in protein synthesis in rat vascular smooth muscle cells Serum amyloid A induces NLRP-3-mediated IL-1β secretion in neutrophils Neutrophil cell surface receptors and their intracellular signal transduction pathways Oxidative stress is a critical mediator of the angiotensin II signal in human neutrophils: involvement of mitogen-activated protein kinase, calcineurin, and the transcription factor NF-κB Rac2 GTPase activation by angiotensin II is modulated by Ca2+/calcineurin and mitogen-activated protein kinases in human neutrophils Phosphorylation of the adaptor ASC acts as a molecular switch that controls the formation of speck-like aggregates and inflammasome activity Necroptosis molecular mechanisms: recent findings regarding novel necroptosis regulators Initiation and execution mechanisms of necroptosis: an overview Necroptosis controls NET generation and mediates complement activation, endothelial damage, and autoimmune vasculitis The pseudokinase MLKL activates PAD4-dependent NET formation in necroptotic neutrophils Angiotensin II triggers RIPK3-MLKL-mediated necroptosis by activating the Fas/FasL signaling pathway in renal tubular cells Cell-Cycle proteins control production of neutrophil extracellular traps An extracellular signal-regulated kinase 1-and 2-dependent program of chromatin trafficking of c-Fos and Fra-1 is required for cyclin D1 expression during cell cycle reentry Angiotensin II-induced overexpression of sirtuin 1 contributes to enhanced expression of Giα proteins and hyperproliferation of vascular smooth muscle cells Stimulation of NADPH oxidase by angiotensin II in human neutrophils is mediated by ERK, p38 MAP-kinase and cytosolic phospholipase A2 Angiotensin II activation of cyclin D1-dependent kinase activity Angiotensin II and serum differentially regulate expression of cyclins, activity of cyclin-dependent kinases, and phosphorylation of retinoblastoma gene product in neonatal cardiac myocytes Low-density granulocytes: functionally distinct, immature neutrophils in rheumatoid arthritis with altered properties and defective TNF signaling Nuclear envelope rupture and NET formation is driven by PKCα-mediated lamin B disassembly Phosphorylation of lamin B at the nuclear membrane by activated protein kinase C Oxidative burstdependent NETosis is implicated in the resolution of necrosis-associated sterile inflammation PI3Kγ controls oxidative bursts in neutrophils via interactions with PKCα and p47phox Impaired NADPH oxidase activity in Rac2-deficient murine neutrophils does not result from defective translocation of p47phox and p67phox and can be rescued by exogenous arachidonic acid Zymosan induces NADPH oxidase activation in human neutrophils by inducing the phosphorylation of p47phox and the activation of Rac2: involvement of protein tyrosine kinases, PI3Kinase, PKC, ERK1/2 and p38MAPkinase Rac2 is required for the formation of neutrophil extracellular traps A key role for Rac and Pak signaling in neutrophil extracellular traps (NETs) formation defines a new potential therapeutic target 2-Chlorofatty acids: lipid mediators of neutrophil extracellular trap formation A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis Effects of angiotensin II receptor blockers and ACE (angiotensinconverting enzyme) inhibitors on virus infection, inflammatory status, and clinical outcomes in patients With COVID-19 and hypertension: a single-center retrospective study Antecedent administration of angiotensin-converting enzyme inhibitors or angiotensin II Receptor antagonists and survival after hospitalization for COVID-19 syndrome Renin-angiotensin system blockers and the COVID-19 pandemic: at present there is no evidence to abandon reninangiotensin system blockers Angiotensin II type-1 receptor antagonism attenuates the inflammatory and thrombogenic responses to hypercholesterolemia in venules Thromboplasminflammation in COVID-19 coagulopathy: three viewpoints for diagnostic and therapeutic strategies Efficacy and mechanism of angiotensin II receptor blocker treatment in experimental abdominal aortic aneurysms Long-term renin-angiotensin blocking therapy in hypertensive patients with normal aorta may attenuate the formation of abdominal aortic aneurysms Angiotensinconverting enzyme 2, coronavirus disease 2019, and abdominal aortic aneurysms Type-4 phosphodiesterase (PDE4) blockade reduces NETosis in cystic fibrosis Prostaglandin E2 inhibits neutrophil extracellular trap formation through production of cyclic AMP Rodríguez, Rolipram Prevents the Formation of Abdominal Aortic Aneurysm (AAA) in Mice: PDE4B as a Target in AAA Letter to the editor: immunomodulation by phosphodiesterase-4 inhibitor in COVID-19 patients The protein kinase A negatively regulates reactive oxygen species production by phosphorylating gp91phox/ NOX2 in human neutrophils, Free Radic Protein kinase A downregulates the phosphorylation of p47 phox in human neutrophils: a possible pathway for inhibition of the respiratory burst This work was supported by grants from the Specialized Research Fund for the National Natural Science Foundation of China (81973511).