Skip to main content
American Journal of Physiology - Cell Physiology logoLink to American Journal of Physiology - Cell Physiology
. 2020 Jul 15;319(3):C510–C532. doi: 10.1152/ajpcell.00181.2020

Neutrophil heterogeneity and fate in inflamed tissues: implications for the resolution of inflammation

János G Filep 1,, Amiram Ariel 2
PMCID: PMC7509268  PMID: 32667864

Abstract

Neutrophils are polymorphonuclear leukocytes that play a central role in host defense against infection and tissue injury. They are rapidly recruited to the inflamed site and execute a variety of functions to clear invading pathogens and damaged cells. However, many of their defense mechanisms are capable of inflicting collateral tissue damage. Neutrophil-driven inflammation is a unifying mechanism underlying many common diseases. Efficient removal of neutrophils from inflammatory loci is critical for timely resolution of inflammation and return to homeostasis. Accumulating evidence challenges the classical view that neutrophils represent a homogeneous population and that halting neutrophil influx is sufficient to explain their rapid decline within inflamed loci during the resolution of protective inflammation. Hence, understanding the mechanisms that govern neutrophil functions and their removal from the inflammatory locus is critical for minimizing damage to the surrounding tissue and for return to homeostasis. In this review, we briefly address recent advances in characterizing neutrophil phenotypic and functional heterogeneity and the molecular mechanisms that determine the fate of neutrophils within inflammatory loci and the outcome of the inflammatory response. We also discuss how these mechanisms may be harnessed as potential therapeutic targets to facilitate resolution of inflammation.

Keywords: cell death, efferocytosis, neutrophil trafficking, neutrophils, specialized proresolving mediators

INTRODUCTION

Acute inflammation is a localized, self-limited innate host defense mechanism against invading pathogens and tissue injury. Typically, polymorphonuclear neutrophil granulocytes (PMN) are among the first cells to be recruited from the blood to the site of infection or injury. Neutrophils possess an impressive array of mechanisms that contribute to elimination of offending agents and necrotic tissues (93, 199). Excessive or dysregulated neutrophil recruitment and activation are also capable of inflicting tissue damage and prolong inflammation (93, 134, 197). Preclinical data indicate that impaired neutrophil removal from inflamed tissues results in aggravation and prolongation of the inflammatory responses (197). Neutrophil-driven inflammation is a common mechanism underlying many pathological conditions, including cardiovascular, acute respiratory, neurodegenerative, metabolic, and autoimmune diseases, sepsis, asthma, and cancer (136, 159). Hence, efficient inactivation and removal of emigrated neutrophils is critical for timely resolution of inflammation and return to homeostasis.

The molecular events governing the multiple-step process of neutrophil trafficking into tissues have been well defined (136, 157, 205). However, the fates of emigrated neutrophils have received considerably less attention. This is likely based on the long-held view that halting influx of additional neutrophils and clearance of neutrophils that fulfilled their immediate mission in the tissues can explain their rapid decline in inflamed areas and return to homeostasis. This review will focus on recent advances in molecular events that determine the fate of neutrophils that have emigrated into the inflamed and injured tissue and discuss how these mechanisms may be harnessed as potential therapeutic targets of the future.

NEUTROPHIL HETEROGENEITY

Phenotypical and functional heterogeneity is well known for lymphocyte and macrophage populations and assures diverse functions in homeostasis, inflammation, or immunity (90, 311). In contrast, neutrophils are traditionally considered as a homogeneous population of terminally differentiated cells with highly conserved function. Human blood neutrophils have banded or segmented nuclei, contain heterogeneous cytoplasmic granules, and express Mac-1(CD11b), CD15, CD16/Fcγ receptor III, CD33, and CD66b, which along with lack of CD14 expression are widely used to distinguish PMN from eosinophils and monocytes. However, emerging evidence challenges the classical view of PMN homogeneity. In healthy subjects, subsets of neutrophils expressing the glycoprotein CD177 (18, 112), olfactomedin-4 (45, 162), or variable T cell receptor-like immune receptors (84, 230) have been identified. CD177 is required for surface presentation of proteinase 3 (18), a serine protease normally located in the primary granules, which mediates transendothelial migration of CD177+neutrophils (144). Proteinase 3 is a major antigen in anti-neutrophil cytoplasmic antibody (ANCA)-associated systemic vasculitis (275). Patients with ANCA-associated vasculitis have higher number of CD177/proteinase 3-positive neutrophils (113) that may also predict relapse (232). The functional properties of this neutrophil subset are controversial (113, 260), and genetic deletion of CD177 did not affect neutrophil migration in mice (300). Hence, defining the roles of CD177+ and CD177 subsets will require further investigations. Olfactomedin-4, expressed in only 20–25% of human peripheral blood neutrophils (45), does not affect PMN influx into tissues (294), whereas it enhances bactericidal capacity against Staphylococcus aureus in mice (162). A small subset (3–5%) of human neutrophils expresses variable T cell receptor-like immune receptors (230), with declining repertoire diversity in the elderly (84). The function of these receptors is still unknown.

Bone marrow PMNs are derived from the granulocyte-macrophage progenitor, leading to generation of a committed neutrophil precursor (preNeu), which differentiates into nonproliferating immature and mature PMNs (72). These subsets possess distinct proliferative capacity and genetic and functional signatures (72). PreNeus expand in the bone marrow and spleen during inflammation, indicating increased intra- and extramedullary granulopoiesis (72). Unlike preNeus, immature neutrophils could enter the bloodstream and traffic into the inflamed site as efficiently as mature PMNs (72). The implications of mobilization of immature neutrophils to host defense or contribution to PMN functional heterogeneity are currently unclear.

During the acute-phase response, bone marrow neutrophils are mobilized to the blood and consequently to highly vascularized organs (136), which are governed by the cytokine G-CSF, the chemokines CXCL1 and CXCL2 (63, 252), and lactate released from inflammatory bone marrow neutrophils (132). Of note, CXCL1 and CXCL2 are also involved in enhancing different modes of neutrophil transendothelial migration (91). Chemokines acting via CXCR4 may control neutrophil return to bone marrow following senescence (174). CXCR4 expression is almost undetectable on circulating human neutrophils, whereas it is upregulated following ex vivo culture parallel with enhanced migratory capacity toward CXCL12 (194), implying homing to the bone marrow. In mice, upregulation of CXCR4 and CD11b expression and reduction of CD62L expression on circulating PMN defines a neutrophil subset named “senescent” or “aged” neutrophils, which are controlled by circadian oscillations in the number of hematopoietic stem progenitor cells (35), the microbiome (308), and a cell-autologous molecular program consisting of clock-related genes, such as Bmal1, and the CXCR2 signaling pathway (3). The population of “aged” neutrophils (CD62Llow) peaks during daylight, whereas nonaged PMNs (CD62Lhigh) are most prevalent in the circulation at night (3, 35). “Aging” has been suggested to favor PMN clearance (3), whereas other studies reported rapid recruitment of “aged” PMN to sites of inflammation (282). Circadian changes in CD62L and CXCR2 expression were also detected in human PMN, suggesting conservation of this phenomenon across species (3) and endorsing time-based therapeutic interventions for optimizing clinical benefit (236).

Several studies have identified intrinsic functional heterogeneity in human circulating neutrophil pool under physiological conditions (226). For example, preference of some neutrophils over others to phagocytose bacteria, termed as competitive phagocytosis, has been reported (109). However, analysis of common PMN markers failed to identify the phenotype associated with phagocytic function, highlighting the challenges in linking functional responses to phenotype.

In the inflammatory milieu, neutrophils sense and respond to cues from both pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) (22). Neutrophils integrate these signals, which in turn alter their lifespan and capacity to undergo phenotypic and functional changes, and metabolic and transcriptional reprogramming, hence, setting the foundation for PMN heterogeneity under pathological conditions (21, 143, 260). Not surprisingly, a number of PMN subsets have been reported. For instance, LPS administration to healthy volunteers led to increases in the number of circulating banded CD16dim neutrophils (as opposed to mature fully competent segmented CD16bright neutrophils) with impaired antimicrobial function (225) and mature hypersegmented neutrophils that can suppress T cell responses and proliferation, resembling the actions of myeloid-derived suppressor cells (224). A CD63+ neutrophil subset was isolated from the sputum of patients with cystic fibrosis (115, 277). These neutrophils exhibit increased neutrophil elastase and arginase 1 activity and suppress T cell function, thereby likely contributing to perpetuation and progression of the disease. Other studies have shown that IL-13+ neutrophils polarize macrophages toward M2 phenotype that accelerate clearance of nematodes in mice (37). A portion of circulating neutrophils express IL-17 upon stimulation with IL-6 and IL-23 both in humans and mice, parallel with increased capacity for ROS production and fungal killing (273).

Human neutrophils exposed to PGE2 or PGD2 induce switching from LTB4 synthesis to production of lipoxin A4, a potent stop signal for neutrophil trafficking (155). This lipid class-switching may define a neutrophil phenotype in the resolution phase of inflammation, for lipoxin A4 also facilitates macrophage efferocytosis (92, 253).

Another PMN subpopulation commonly referred to as low-density neutrophils may exert immunosuppressive function (also known as granulocytic myeloid-derived suppressor cells or G-MDSC) or display proinflammatory properties (34). Low-density neutrophils have been detected in healthy pregnant women (137, 265) as well as in patients with cancer (193, 240), sepsis (118), diabetes (298), and autoimmune diseases, such as systemic lupus erythematosus and psoriasis (34, 99). Proinflammatory subsets of low-density neutrophils produce IFNα upon stimulation, display reduced phagocytic activity, and are susceptible to release neutrophil extracellular traps (NET), thereby causing injury to endothelial cells (53, 285). These actions are consistent with aggravation and progression of the clinical symptoms in lupus. Recently, CD10 was suggested as a marker to discriminating immune-suppressive mature (defined as CD66b+, CD10+) and immune-stimulatory immature (CD66b+, CD10) neutrophils present within heterogeneous PMN populations in patients with acute or chronic inflammatory conditions (173). Elevated CD10 and CD66b expression on oral PMN from patients with chronic periodontitis identifies a proinflammatory neutrophil subset distinct from naïve circulatory neutrophils and parainflammatory PMNs, which are present in the healthy oral cavity and interact with commensal biofilms without inducing clinically evident inflammation (75).

Studies on tumor-infiltrating PMNs in mice revealed distinct subsets of neutrophils that play opposing immune-modulator roles (80, 226). A PMN population, referred to as N1, is characterized by hypersegmented nuclei, production of proinflammatory cytokines, and potent tumor killing capacity (80). By contrast, the N2 subset displays an immature phenotype and increased arginase activity and favors tumor growth (80). Immature PMNs, defined as CD101 cells (as opposed to CD101+ mature PMNs), were found to accumulate at the tumor site and correlate with cancer progression (72). It is still unclear whether the N1 and N2 populations share a common origin or are originated from naïve mature neutrophils and G-MDSC, respectively (81, 240). Interestingly, the anti-inflammatory cytokine transforming growth factor-β (298) and IFNβ (6, 116) have been implied in polarizing tumor-associated neutrophils toward an anti-angiogenic and anti-tumorigenic phenotype. Other studies have shown that expression of the proto-oncogene Met is essential for recruitment of anti-tumor neutrophils (76). A unique subset of tumor-associated PMNs has been identified in early-stage (small size) human lung cancer (261). This subset possesses characteristics of both neutrophils and antigen-presenting cells and could trigger an anti-tumor T cell response. Intriguingly, these cells were completely absent in larger tumors (261), indicating the phenotypical and functional divergence of tumor-associated PMNs during human lung cancer progression (241).

MULTIDIRECTIONAL MIGRATION

Neutrophils are usually the first cells to accumulate in injured and microbe-infected tissues (93, 199). Neutrophils egress from the blood to tissues is a multistep process involving PMN activation, rolling on, and adhesion to the activated endothelium, followed by transendothelial migration and chemotactic migration toward the inflammatory locus. The molecular mechanisms underlying and governing this process have been described in detail (136, 157, 205). The acute inflammatory response is protective and self-limited and resolves on its own under healthy conditions. Proteins such as annexin A1 (218, 262) and galectin-1 (150, 301) and proresolving lipids, including lipoxins, resolvins, protectins, and maresins, are produced during the resolution phase of self-limited inflammation (155, 253, 254) and function as potent stop signals for PMN activation and trafficking into tissues (218, 253, 257). Blocking PMN recruitment is an important event in terminating the inflammatory reaction for uncontrolled, excessive neutrophil accumulation and can perpetuate tissue damage that aggravates the initial protective inflammatory response and may evolve into chronic inflammation. Figure 1 summarizes neutrophil mobilization into sites of infection or tissue injury and PMN functions in host defense.

Fig. 1.

Fig. 1.

Neutrophil kinetics and fate of neutrophils in inflamed tissues. Neutrophils are rapidly recruited from the blood to infected or injured tissues. Neutrophil trafficking is a tightly regulated multistep process. Mobilization of annexin A1 to the neutrophil surface results in detachment of adherent polymorphonuclear neutrophil granulocytes (PMNs), thereby limiting their recruitment. Following extravasation, PMNs swarm toward and form clusters around the affected site. Neutrophils may neutralize and clear bacteria and damaged cells through phagocytosis, release of net extracellular traps (NETs), or necroptosis. Phagocytosis induces apoptosis in PMNs, followed by removal of apoptotic cells by macrophages through efferocytosis. Efferocytosis induces polarization of macrophages toward proresolution phenotype. NETs immobilize and kill bacteria in the extracellular milieu and may also present autoantigens to trigger adaptive immunity. Under certain conditions, PMNs can undergo necroptosis (programmed necrosis) parallel with or independent from NET formation. Necrotic debris may then be cleared by macrophages. Although several molecular switches that govern neutrophil responses to bacteria have been identified, the signals that direct the choice between phagocytosis, NET formation, and necroptosis are still poorly characterized. Emigrated PMN may reenter the circulation through reverse transendothelial migration (TEM) or exit from inflamed tissues through the lymphatic vessels. These “recirculated” PMNs may carry and present antigens to T and B cells in the lymph nodes or return to bone marrow for degradation. CRAMP, cathelin-related antimicrobial peptide; DAMPs, damage-associated molecular patterns; GSDM D, gasdermin D; JAM-C, junctional adhesion molecule-C; LTB4, leukotriene B4; LXA4, lipoxin A4; NE, neutrophil elastase; PAMPs, pathogen-associated molecular patterns; ROS, reactive oxygen species; RvD1, resolvin D1; RvE1, resolvin E1.

Neutrophil Detachment

According to the classical concept of neutrophil trafficking, adhesion of PMNs to endothelial cells is an essential step to direct them to sites of inflammation. Advances in imaging techniques have revealed that adherent neutrophils can detach and move away from sites of inflammation. Detachment of adherent PMNs is mediated by increased production and secretion of annexin A1 in response to glucocorticoids (171, 218), mast cell-stabilizing anti-allergic drugs (302), natural serine protease inhibitors (284), and the gaseous mediator hydrogen sulfide (27). Annexin A1 is mobilized from the cytoplasm pool to cell surface and signals through the lipoxin A4/formyl-peptide receptor 2 (ALX/FPR2) (217, 218). Lipoxin A4 evokes mobilization of annexin A1 (26) and acts in concert with annexin A1, thus forming an endogenous anti-inflammation loop to limit PMN trafficking into inflammatory loci (26). Interestingly, a small portion of neutrophils continued to pass the endothelial barrier following treatment with glucocorticoids (171), likely representing a functionally distinct subpopulation. The potential importance of annexin A1 controlling neutrophil adhesion is supported by the findings of enhanced neutrophil transmigration in annexin A1-deficient mice (36). However, the phenotype, function, and fate of detached neutrophils remain to be investigated.

Neutrophil Swarming

Following extravasation from the blood, PMN behavior can switch from mere chemotaxis toward the infected or injured site to a swarm-like migration pattern, referred to as neutrophil swarming (148, 202). Neutrophil swarming is characterized by sequential phases of highly coordinated interstitial chemotaxis and progressively accelerated exponential PMN accumulation, culminating in the formation of neutrophil clusters around the damage core to insulate the affected site from the surrounding healthy tissues (43, 133, 147, 148). Neutrophil swarms require LTB4 released from PMNs (4, 148). The signal(s) for enhanced LTB4 formation has not been elucidated, although contact of PMNs with necrotic cells is critical for initiation of swarming (281). It is possible that upon sensing necrotic tissue, pioneer neutrophils release ATP through connexin 43 channels, which in turn opens P2X1 channels, leading to elevations in intracellular calcium and subsequent activation of 5-lipoxygnease and LTB4 production (228). Coordinated activation of LTB4 synthesis would then build a stable LTB4 gradient to drive concerted waves of migration. Consequent microscale arrays confirmed the dominant role of LTB4 during human neutrophil swarming and also identified a large number of protein mediators that function to enhance neutrophil accumulation during swarming (235). Among these proteins are CXCL8, galectin-3, CXCL7, lipocalin-2, and pentraxin-3. The integrins Mac-1and LFA-1 are critical for PMN accumulation in the collagen-free injury center (148). Signaling through CXCR1, CXCR2, ALX/FPR2, and LTB4 receptor BLT1 coordinates neutrophil congregation at damaged sites and maintains tight neutrophil association in the aggregation stage (49, 148). These findings identify a feedforward loop centered on LTB4 in both the recruitment and the clustering phases of swarming. By cloaking the injured area (removing debris and sequestering DAMPs), tissue-resident macrophages can prevent swarming and consequently maintain tissue functionality (281). However, because cloaking did not interfere with swarm progression after its initiation and PMNs still appeared around cloaked areas, it is unlikely that macrophage inhibition of swarming was mediated by secretion of chemorepulsive signals (281). Moreover, development of human neutrophil swarms was associated with generation of the potent proresolving lipid mediators lipoxin A4 and resolvin E3 parallel with slower growth and reduced size of swarms (235).

Neutrophil swarming has been observed in vitro around dying cells (169) and in animal models of sterile inflammation (148, 281), infection with a wide range of pathogens, including Staphylococcus aureus (124), Escherichia coli (140), Toxoplasma gondii (43), and Aspergillus fumigatus (140), and following influenza vaccination (227). Under these conditions, swarming was found to limit the extent of tissue damage and infection, implying an important role in host defense. Consistently, neutrophils isolated from trauma patients (235) or from patients receiving immunosuppressive therapy following transplantation (119) were shown to have a limited ability to swarm. Thus, impaired neutrophil swarming may contribute to increased susceptibility and inability to clear Staphylococcus infections following major trauma (154) and fungal infections with Aspergillus fumigatus conidia in patients during immunosuppressive therapy (119). Excessive or uncontrolled swarms may contribute to aggravation and propagation of tissue damage, as was shown in bacteria and ischemia-reperfusion-induced pulmonary inflammation in mice (140) and inflammation flares around uric acid crystals in gout disease (170).

Reverse Transendothelial Migration

Studies using intravital microscopy documented the ability of neutrophils to exhibit motility away from injured tissues back into the vascular lumen, known as reverse transendothelial migration (TEM) (46, 146, 299). This phenomenon was first demonstrated using in vitro models of neutrophil transmigration (30) and subsequently in zebrafish embryos (176) and in the mouse cremaster circulation (299). Neutrophil reverse TEM is most prevalent in tissues subjected to ischemia-reperfusion injury that is associated with reduced expression of junctional adhesion molecule-C (JAM-C) at endothelial cell junctions (246, 299). Pharmacological blockade or genetic deletion of JAM-C enhances the frequency of neutrophil reverse TEM in mouse cremaster venules (299). Under ischemia-reperfusion, excessive production of LTB4 leads to activation of the LTB4 receptor BLT1 on neutrophils and release of neutrophil elastase (46). Neutrophil elastase binds to Mac-1 (32), which is also a ligand for JAM-C and cleaves JAM-C (46). In vitro, reversely migrated neutrophils display a phenotype (ICAM-1high, CXCR1low) distinct from tissue-resident (ICAM-1low, CXCR1low) or circulating (ICAM-1low, CXCR1high) neutrophils and increased capacity to produce superoxide (30, 299). ICAM-1high neutrophils were also detected in both the systemic and pulmonary circulation of mice subjected to either cremaster muscle or lower-limb ischemia-reperfusion injury (299). In the cremaster model, an association was found between percentage of ICAM-1high neutrophils within the pulmonary vasculature and the severity of lung inflammation (299).

At present, the functional implication of this phenomenon is unclear. One possibility is that reverse TEM serves as a protective mechanism to enhance clearance of PMNs from infected or injured tissues, thereby contributing to dampening the inflammatory response (176, 206). Alternatively, neutrophil reverse TEM may be associated with dissemination of systemic inflammation and distant organ injury (46, 299).

Lymphatic Transit

Neutrophils may also exit from the inflamed tissue through the lymphatic vessels. Neutrophil accumulation in draining lymph nodes in mice has been detected following injection of a variety of bacteria, protozoan parasites, and viruses (1, 43, 102, 103, 148). Neutrophils have been observed in the subcapsular sinus, medullary region, interfollicular zone, and T cell zone, depending on the pathogen and the mode of PMN recruitment (103). Neutrophil localization within the lymph node affects its function. For instance, rapid localization of neutrophils to the subcapsular sinus can kill S. aureus and destroy infected cells (124), thereby limiting further pathogen spread. In the interfollicular zone, neutrophils are positioned in close proximity to T and B cells and innate-like lymphocytes such as NK cells (124, 127) and are required for optimal NK cell activity (117). Some studies have reported that PMNs can present antigen to T cells in vitro (271). However, it is not known whether lymph node neutrophils can present antigen to T cells in vivo.

Neutrophils may enter lymph nodes from the bloodstream through high endothelial venules or lymphatic vessels (25, 124). The surface molecules governing PMN transmigration across high endothelial venules are relatively well defined and include CD62L, CD62P, CD162, LFA1 (CD11a), Mac-1 (CD11b), ICAM-1, and CXCR4 (103). Studies with CXCR2-deficient mice showed reduced neutrophil influx to lymph nodes in a tumor lysis model (25), whereas CXCR2 was dispensable for Staphylococcus aureus-induced PMN accumulation in lymph nodes (102). The mechanisms governing neutrophil migration via lymphatic vessels are still being investigated. Neutrophil migration from inflamed skin to draining lymph nodes is guided by CXCR4, and its ligand CXCL12 expressed by lymphatic endothelial cells (102). These findings would imply CXCR4 as a potential therapeutic target to reduce neutrophil entry into lymphatic vessels. Other studies suggest that Mac-1 may also be required for neutrophil entry into lymphatics following intradermal injection of Mycobacterium bovis in mice (237). The counter-ligand(s) for Mac-1-dependent migration has not yet been identified.

Staphylococcus aureus-pulsed neutrophils that migrated to the sentinel lymph nodes exhibit a distinct phenotype (characterized as CD11bhigh, CD62Llow, and CXCR2low) and upregulation of major histocompatibility complex (MHC) II and the costimulatory molecules CD80 and CD86, implying their ability of antigen presentation and initiation of adaptive immunity (102). Moreover, PMNs from vaccinia virus-infected skin were reported to migrate to the bone marrow and enhance activity of virus-specific memory CD8+ (cytotoxic) T cells (60). Collectively, these observations would indicate that neutrophil-associated antigen transport could contribute to trigger adaptive immunity. By contrast, other studies have shown that neutrophils carrying Leishmania major (221) or Toxoplasma gondii (48) contribute to immune evasion and pathogen spread throughout the mouse intestine.

NEUTROPHIL LIFESPAN

Human circulating neutrophils have a short half-life (145, 270), although one study estimated their lifespan to be 5.4 days (223). In the inflammatory microenvironment, neutrophils receive opposing cues that modulate their lifespan and their mode of death. Cell death is a major mechanism to eliminate neutrophils from inflammatory loci for assuring timely resolution of inflammation. Neutrophils are classically thought to die via apoptosis (244). Apoptosis is a form of programmed cell death that involves cell shrinkage and chromatin condensation, followed by fragmentation of the entire cell into sealed apoptotic bodies. Execution of this constitutively expressed cell death program renders neutrophils unresponsive to inflammatory stimuli (135) and promotes their removal via efferocytosis (89, 138, 243) with minimal or no damage to the surrounding tissue. Although PMN apoptosis has been studied most extensively, neutrophil subsets undergo other modalities of caspase-independent regulated cell death, such as NETosis (82) and necroptosis (213). Necrotic cell death can be accidental or programmed (i.e., necroptosis) and is characterized by loss of plasma membrane integrity, release of intracellular contents into the extracellular space, and proinflammatory actions. Moreover, if apoptotic neutrophils are not cleared in a timely fashion, they may undergo secondary necrosis. Release of neutrophil constituents may contribute to killing bacteria (28) or to sustain collateral tissue damage and trigger autoimmunity (98, 263). Thus, the mechanisms activated by emigrated PMNs to contain pathogens or tissue damage through phagocytosis, necrosis, or NET formation would ultimately determine their own fate, the means of clearance of debris generated by apoptosis versus necrosis and ultimately the outcome of the inflammatory response.

Apoptosis and Phagocytosis-Induced Cell Death

Neutrophil lifespan is thought to be extended within inflammatory loci. PMN adhesion to endothelial cells and most inflammatory mediators and microbial constituents found in the inflammatory microenvironment generate prosurvival cues to prolong neutrophil longevity by delaying the constitutive apoptotic program (47, 122, 152). Activated neutrophils secrete protons that contribute to interstitial acidification, yielding pH between 5.5 and 7.0, in inflammatory loci (64, 88, 195). Extracellular acidosis is a danger signal (231), which through activation of multiple signaling pathways, including ERK 1/2, phosphoinositide 3-kinase (PI3K)/Akt, NF-κB, and cAMP/PKA leads to preventing degradation of Mcl-1, a key regulator of neutrophil survival (62), and subsequently suppresses PMN apoptosis and enhances the apoptosis-delaying actions of inflammatory mediators (65). Extracellular acidosis affects the outcome of inflammation in a context-dependent manner. For example, genetic deletion of the pH sensor TDAG8 aggravates LPS-induced acute lung injury in mice (279), whereas proton pump inhibitors protect mice from the deleterious actions of persisting acidosis in acute systemic inflammation (14). Acidosis is a characteristic feature of patients with sepsis, and more severe acidosis is associated with poor prognosis (203). Hypoxia in the inflammatory microenvironment and bacterial infections even under normoxia can induce release of HIF-1α (222) and HIF-2α (276), which activate prosurvival mechanisms in neutrophils, control their bactericidal activity, and restrict systemic spread of infection. Moreover, activated neutrophils release myeloperoxidase, which has been implicated in killing bacteria and tissue destruction (134, 198), binds to Mac-1 on human neutrophils (149), induces PMN activation and degranulation (68, 149), and delays intrinsic apoptosis (68). Thus, myeloperoxidase triggers a feedforward loop to amplify and perpetuate the inflammatory response (68). Accordingly, myeloperoxidase prolongs neutrophil-mediated acute lung injury in mice (68). Genetic deletion of myeloperoxidase in mice attenuates PMN accumulation and organ damage after renal ischemia-reperfusion (177) and reduces E. coli septicemia-induced lung injury and mortality (29). Conversely, aspirin-triggered 15-epi-LXA4 interrupts the myeloperoxidase-centered circuit and redirects neutrophils to apoptosis (69).

Neutrophil apoptosis has emerged as one of the critical determinants of the outcome of the inflammatory response. Extended neutrophil lifespan through delayed apoptosis is observed in patients with sepsis (128), acute respiratory distress syndrome (178), severe asthma (280), or acute coronary artery disease (87) and is associated with disease progression and poor prognosis. Moreover, neutrophil heterogeneity is a common finding in the bronchial lavage of patients with chronic obstructive airway diseases, where the number of hypersegmented PMNs inversely correlates with lung function (163). In experimental models, delaying neutrophil apoptosis adversely affects the duration of inflammation (68, 120), whereas treatment with cyclin-dependent kinase inhibitors, 15-epi-LXA4, or IFN-β counters prosurvival cues, facilitates neutrophil apoptosis and efferocytosis, and consequently enhances resolution of inflammation (69, 238, 242). Genetic deletion of the apoptosis-related effector protein in the TGFβ signaling pathway (ARTS/Sept4_i2) prolongs inflammation in mice (168), underscoring the importance of the intrinsic (mitochondrial) pathway of apoptosis in the resolution of inflammation.

Phagocytosis-Induced Cell Death

Typically, phagocytosis of opsonized bacteria or necrotic cells accelerates neutrophil apoptosis and facilitates the removal of apoptotic neutrophils via efferocytosis. Lateral clustering of Mac-1 [or complement receptor 3 (CR3)] initiates complement-mediated phagocytosis (79), which is governed by a delicate balance between Mac-1 and the complement C5a receptor (C5aR or CD88) (97, 187). Diminished expression of CXCR1 or Mac-1 on neutrophils results in reduced phagocytosis of bacteria (20, 105). Pharmacological blockade or genetic deletion of C5aR reduces phagocytosis of E. coli and P. aeruginosa by human and mouse neutrophils, respectively (187). The PAMP bacterial DNA and the danger-associated signal mitochondrial DNA, sharing the bacterial ancestry, reduces phagocytosis and phagocytosis-induced apoptosis in human neutrophils through neutrophil elastase and proteinase 3-mediated shedding of C5aR (Fig. 2) (250). Consistently, bacterial DNA delays clearance of E. coli, neutrophil apoptosis, and efferocytosis and prolongs E. coli-evoked lung injury in mice (250). TLR9-mediated cleavage of C5aR may represent a potential strategy evolved by bacteria to avoid complement-mediated destruction (146) and may also explain defects in neutrophil function, including impaired phagocytosis under pathological conditions characterized by release of bacterial DNA (233, 249) or mitochondrial DNA (295, 309). Aspirin triggered 15-epi-LXA4 and 17-epi-RvD1, acting through the ALX/FPR2, effectively counter TLR9 signaling, prevent release of neutrophil elastase and proteinase 3, and restore the balance between Mac-1 and C5aR expression, phagocytosis, and phagocytosis-induced apoptosis in human PMNs (250). The therapeutic potential of these lipids is illustrated by the observations of accelerated bacterial clearance, enhanced neutrophil apoptosis, and efferocytosis in a mouse model of acute lung injury (250). Of note is that other proresolving lipids, such as resolvin E1, acting through the LTB4 receptor BLT1 (66), and resolvin D5, which signals through GPR32 (39), facilitate phagocytosis by naïve PMNs via distinct receptors and molecular mechanisms.

Fig. 2.

Fig. 2.

Modulation of neutrophil phagocytosis and phagocytosis-induced cell death. Typically, Mac-1-mediated phagocytosis of opsonized bacteria or cell debris leads to polymorphonuclear neutrophil granulocytes (PMN) apoptosis by reactive oxygen species (ROS)-dependent activation of caspase-8. Ligation of Toll-like receptor 9 (TLR9) by bacterial DNA (CpG DNA) or mitochondrial DNA (mtDNA) released from necrotic cells evokes release of contents of azurophil granules. Neutrophil elastase (NE) and proteinase 3 (PR3) downregulate complement 5a receptor (C5aR) and by altering the Mac-1/C5aR ratio reduce phagocytosis, bacterial clearance, and phagocytosis-induced cell death (PICD). Myeloperoxidase (MPO) binds to Mac-1 and generates survival signals for PMNs, consistent with prolongation of inflammation. Specialized proresolving lipid mediators efficiently reverse these actions (right). Resolvin E1 (RvE1), acting through the leukotriene B4 receptor (BLT1), enhances phagocytosis, whereas aspirin-triggered 15-epi-LXA4 (lipoxin A4) or 17-epi-RvD1 (resolvin D1), through lipoxin A4/formyl-peptide receptor 2 (ALX/FPR2), inhibits TLR9-stimulated degranulation, restores Mac-1/C5aR ratio, phagocytosis, and bacterial killing, and counters MPO-generated survival signals, thereby facilitating resolution. Hck, hematopoietic cell kinase.

Receptors Integrating Life and Death Decisions

Outside-in signaling through β2-integrins.

The β2-integrin Mac-1 is best known for mediating neutrophil adherence to endothelial cells and the extracellular matrix (136, 157) and phagocytosis of complement C3b-opsonized bacteria (61) as well as mediating phosphatidylserine- and C3b-dependent efferocytosis by human macrophages (186). The β2-integrins function as bidirectional “signaling machines” (114) that also regulate the neutrophil lifespan. Engagement of Mac-1 with its endothelial counterreceptor ICAM-1, fibrinogen, immune complexes, platelets, or myeloperoxidase induces activation of the PI3K/Akt and MAPK/ERK signaling pathways, leading to Mcl-1 accumulation and suppression of the intrinsic apoptotic program (68, 182). Mac-1 ligation in conjunction with TNF-α or FAS ligand directs neutrophils toward apoptosis though induction of NADP oxidase/ROS-mediated activation of Lyn and SHIP [Scr-homology 2 (SH2)-containing inositol 5-phosphatase], which inactivates phosphoinositide 3-kinase (182). This mechanism may be relevant for limiting tissue accumulation of neutrophils in vivo. Phagocytosis of C3b-opsonized bacteria induces ROS-dependent activation of caspase-8, which overrides the ERK-mediated survival signal, resulting in apoptosis (66, 182). Caspase-8 forms a heterodimer with FLIP (FLICE-inhibitory protein), which suppresses RIPK3-dependent necroptosis (207, 208), thereby preventing release of neutrophil content.

Downregulation of Mac-1 expression and subsequent modulation of outside-in signaling by proresolving lipid and protein mediators (218, 253, 257) are critical for limiting further PMN recruitment, the lifespan of emigrated neutrophils and neutrophil-mediated tissue damage.

Opposing signaling through formyl peptide receptors.

Formyl peptide receptor (FPR) ligands are ubiquitous in inflammatory loci, are highly diverse functionally, and govern neutrophil trafficking, lifespan, and clearance (303). FPR1, the high-affinity receptor for N-formyl peptides released from bacteria and mitochondria of necrotic cells, is critical for neutrophil killing of bacteria (86, 161, 209), host-commensal interaction during dysbiosis (188), and guiding neutrophils to the necrotic center of focal injury in mice (40, 184).

The lipoxin A4 receptor/formyl-peptide receptor 2 (ALX/FPR2) binds an unusually large number of structurally distinct host- and bacteria-derived agonists, which often evoke opposing biological activities (303). For instance, the acute-phase protein serum amyloid A (SAA) induces neutrophil activation and promotes inflammation (108), whereas LXA4, RvD1 and annexin A1 inhibit neutrophil activation and adhesion to the endothelium, thereby limiting PMN trafficking into tissues (217, 218, 253, 257).

Within inflammatory loci, neutrophils receive and integrate opposing cues from various ALX/FPR2 ligands, and their fate would ultimately depend on the relative abundance of these mediators. For instance, SAA (67) and the antimicrobial peptide LL-37 (287) delay whereas annexin A1 enhances neutrophil apoptosis (220). LXA4 and 15-epi-LXA4, which do not affect PMN apoptosis (123), override the prosurvival signal from SAA, thereby redirecting neutrophils to apoptosis (67). By contrast, SAA produced within the lung opposes LXA4 and contributes to persisting inflammation in patients with chronic obstructive pulmonary disease (COPD) (24). Annexin A1 and RvE1 enhance synthesis of LXA4 (107, 217), forming a feedforward resolution circuit. Ligand selectivity may directly be related to the conformational landscape of FPRs that alters the activation of downstream signaling pathways (50, 74, 142). Thus, LXA4 or annexin A1 stimulate ALX/FPR2 homodimerization and activation of the p38 MAPK-MAPKAPK-Hsp27 pathway to augment the production of the anti-inflammatory cytokine IL-10 (50). Annexin A1-derived peptide Ac2-26 facilitates FPR1-ALX/FPR2 heterodimer formation and activation of JNK and caspase-3, thereby propagating proapoptotic signaling (50, 74). SAA and LL-37 do not induce dimerization; rather, they signal through ALX/FPR2 isomer to activate the Akt and ERK pathways to prevent degradation of Mcl-1 and delay apoptosis (50, 67). Conceivably, abundance of proinflammatory ALX/FPR2 ligands would contribute to the initial phase of the inflammatory response, whereas increased production of proresolving ligands would counter the actions of proinflammatory mediators during the resolution phase.

Efferocytosis

Removal of apoptotic cells is an essential process for development and maintenance of the organism’s health. Efferocytosis is orchestrated by “find me” signals that direct macrophages and dendritic cells toward dying neutrophils and “eat me” signals expressed on the surface of the apoptotic cell, which facilitate their recognition and engulfment (57, 138, 190, 267). Uptake of apoptotic cells is a silent nonphlogistic process that leads to reprogramming of macrophages from the inflammatory phenotype to an anti-inflammatory phenotype (9, 258) and subsequently to a proresolving CD11blow subset with minimal phagocytic activity (31, 242, 247). This latter subset is characterized by expression of 12/15-lipoxygenase and IFN-β-related gene signature. Inhibition of cyclin-dependent kinases 5 and 9 enhances neutrophil apoptosis and accelerates PMN clearance and macrophage reprogramming (111, 238). Phagocytosis of apoptotic neutrophils triggers metabolic switch in engulfing macrophages, leading to glycolysis, lactate release through SLC16A1, and generation of an anti-inflammatory microenvironment (191). Loss of phagocytic capacity in macrophages (when macrophages become satiated) is associated with suppression of glycolysis and increased oxidative phosphorylation (31). Mitochondrial ROS production through the uncoupling protein UCP2 (211), the fission-promoting protein dynamin-related protein (DRP1) (291), and apoptotic corpse-derived l-arginine converted by arginase 1 and ornithine decarboxylase to ornithine and putrescin within the macrophage has been implicated in maintaining efficient efferocytosis. Thus, controlling ROS burden from apoptotic corpses following neutrophil efferocytosis is likely maintained by mitochondrial fission, whereas satiation is associated with arginase 1 shutdown (247).

Apoptotic neutrophils express CCR5, CXCR4, and the atypical chemokine receptor ACKR2/D6 on their surface, which affect the outcome of inflammation (8, 104, 212). CCR5 binds inflammatory chemokines and accelerates their clearance (8), whereas ACKR2/D6 binds CCL5 and enhances reprogramming of resolution phase macrophages (13, 212). Satiated macrophages are a likely source of SPMs such as LXA4, RvD1, and maresin 1 (164, 247). These lipids enhance efferocytosis, macrophage reprogramming, and/or departure from resolving sites (41, 51, 92, 247). Similar properties were reported for protein effectors of resolution, including protein S (164), annexin A1 (51), IL-10 (33), DEL-1 (139), and IFN-β (242). IFN-β seems to play a unique role in the resolution of inflammation, as it is produced by satiated macrophages and possesses multiple proresolving activities, including facilitation of bacterial clearance (125, 242), acceleration of neutrophil apoptosis (7, 242), enhancing efferocytosis, and macrophage reprogramming to the CD11blow phenotype (242) (Fig. 3). Thus, efferocytosis of neutrophils and macrophage reprogramming are tightly controlled by proresolving effectors that are produced following the efferocytic trigger.

Fig. 3.

Fig. 3.

IFN-β regulation of macrophage-neutrophil interactions during resolution. Phagocytosis of bacteria usually induces apoptosis in neutrophils. Polymorphonuclear neutrophil granulocytes (PMN) and tissue resident cells secrete inflammatory mediators that attract blood-borne monocytes to the inflammation site, where they mature to phagocytic inflammatory macrophages that contribute to clearing bacteria and apoptotic PMN. Apoptotic PMN express “find me” signals to attract monocytes and macrophages and “eat me” signals that allow their recognition and engulfment via efferocytosis. Efferocytosis polarizes macrophages from an M1 inflammatory phenotype to an M2 reparative phenotype. Upon reaching efferocytic satiation, M2 macrophages convert to the proresolving Mres phenotype that possesses low phagocytic capacity. Satiated macrophages, and to lesser degree tissue resident macrophages, produce IFN-β, which in turn stimulate bacterial clearance by PMN, redirect neutrophils to apoptosis, and accelerate efferocytosis by resolution phase macrophages. IFN-β reprograms macrophages to the CD11blow phenotype and facilitates their migration to lymphoid organs. In addition to its local actions, IFN-β likely limits excessive tissue repair, fibrosis, bone resorption, and myelopoiesis.

PMN efferocytosis not only governs the responses of the engulfing phagocyte but could also lead to new use of proteins stored in PMN granules. For example, uptake of apoptotic PMN was shown to assist macrophages in limiting the growth of Mycobacterium tuberculosis (272). Lactoferrin, an antibacterial protein stored in the specific granules, was found to accumulate in resolution phase macrophages and undergo fragmentation (165). The lactoferrin fragments released to the extracellular milieu stimulate macrophage reprogramming in vivo, and the formation of resolution-promoting aggregated extracellular traps by human PMN (165).

NET Formation

In addition to phagocytosis and degranulation (i.e., release of preformed antimicrobial peptides, such as myeloperoxidase, neutrophil elastase, or cathelicidin from specific granules), neutrophils can release extracellular traps (NET) to immobilize and kill invading pathogens in the extracellular milieu (16, 28) and to degrade cytokines and chemokines (245). NETs target many pathogens, including Staphylococcus aureus, Mycobacterium tuberculosis, and Aspergillus fumigatus (98), activated platelets and platelet-derived microparticles (172, 180, 286), monosodium urate crystals (245), coagulation factors (175), or microparticles (56, 239). Recent reviews detail the molecular pathways leading to NET formation as well as differences in its composition (121, 210, 263). In general, NETs compromise a DNA scaffold, citrullinated histones, and an array of granule proteins, which absorb pentraxin 3 and complement (70, 307). NET formation, commonly referred to as NETosis, which is considered a form of necrotic cell death (185). Reports also exist of complete release of nuclear DNA, resulting in formation of anuclear but still viable cells (304) and selective extrusion of mitochondrial DNA to form extracellular traps without affecting viability (305). Although differences in the mechanisms responsible for the release of nuclear versus mitochondrial DNA have been proposed (306), the implications of these processes in innate immunity and disease remain to be investigated. Pathways leading to NET formation differ in terms of dependence on ROS (58, 129, 210). Activation of the Raf-MEK-ERK and p38 MAPK pathways triggers NADPH oxidase-dependent release of NETs (100, 130), whereas HMGB1 released from activated platelets, leukocytes, or necrotic cells induces ROS-independent NET formation (269). Because HMGB1 effectively inhibits phagocytosis (15) and acidic environment, generated by H+ ion release during phagocytosis, impairs NET formation (179), these mediators may function as molecular switches to direct the most effective neutrophil response to an insult and represent alternative outcomes of PMN activation. Phagocytosis of bulky phosphatidylserine-exposing particles, such as apoptotic bodies or platelets, renders neutrophils unable to release NETs (172, 181). By contrast, smaller particles or tampering with phosphatidylserine-associated moieties or recognition triggers NETosis and leads to accumulation of apoptotic debris (172). Regardless of the origin of DNA, NETs are degraded directly by DNase 1 (101) released predominantly from dendritic cells (151) or by the cytosolic exonuclease TREX1 (DNase III) following engulfment by macrophages (73, 151). The antimicrobial protein LL-37 protects NET against degradation by bacterial nucleases (246) and is required for the uptake of NET by macrophages (151). Because many of the molecules externalized through NETosis are potent autoantigens, aberrant NET formation and/or impaired NET degradation is recognized as trigger of autoimmune diseases, such as lupus erythematosus and rheumatoid arthritis in susceptible individuals (98).

NET formation is an important mechanism to trap and contain invading pathogens and demarcate the infected areas (44, 83). However, NET formation is increasingly being recognized to contribute to a range of pathologies. For example, clinical studies have reported an association between augmented NET generation and the severity and mortality of pneumonia- or sepsis-associated ARDS (153, 158) and COVID-19-associated ARDS (17, 312). Reducing NET by intratracheal instillation of DNase I or partial deletion of protein deiminase 4 (PAD4+/−) reduced the severity of lung injury and increased survival in a mouse model of severe bacterial pneumonia (153). Intriguingly, complete PAD4 deficiency and attenuated NET formation and lung injury increased bacterial burden, suggesting that bacterial infections may shift the balance of the beneficial and deleterious effects of NET in ARDS. NET formation is also controlled by signaling through ALX/FPR2, for mice with genetic deletion of Fpr2 (the mouse equivalent of human ALX/FPR2) produced excess NETs, and this was associated with more severe lung injury and increased mortality (153).

Necroptosis

Necrotic cell death leads to the release of large amounts of DAMPs and necrotic debris, which are potent inducers of inflammation (213). Recent studies on signaling pathways that are activated in dying neutrophils indicate that under certain conditions PMNs can undergo necroptosis or programmed necrosis, as opposed to necrosis, which occurs in a disorderly manner following cell injury (213). Ligation of TNFR1 in mouse neutrophils leads to activation of the receptor-interacting protein kinase 1 (RIPK1)-RIPK3-mixed lineage kinase domain-like protein (MLKL) signaling and translocation of MLKL1 to the inner leaflet of plasma membrane, leading to membrane permeabilization (297). By ubuquitinilating RIPK1 (293), X-linked IAP (XIAP) functions as a switch to direct neutrophils to either necroptosis or apoptosis (297). It should be noted that high concentrations of TNF-α were required to induce necroptosis in mouse neutrophils, and this phenomenon has not been demonstrated in human neutrophils.

Ligation of adhesion receptors, including CD11b, CD18, CD15, and CD44 in human neutrophils primed with GM-CSF, induces activation of the RIPK3-MLKL-p38 MAPK-PI3K pathway, leading to NADPH oxidase-mediated generation of ROS and subsequent necroptosis (19, 289). In line with an effector role for ROS, neutrophils from patients with chronic granulomatous disease (caused by a genetic defect in NADPH oxidase) do not undergo necroptosis following ligation of adhesion receptors (289). ROS has also been suggested to act upstream of RIPK1-RIPK3-MLKL, thereby triggering necroptosis upon exposure of neutrophils to monosodium urate crystals (54, 55). Consistently, pharmacological blockade of RIPK1 with necrostatin-1 or genetic deletion of RIPK3 reduced neutrophil necroptosis in a mouse model of gouty arthritis (54). In the course of necroptosis, NET formation was also detected in this model (54, 55). However, this could be explained by necrosis-related passive release of chromatin (290), for NET release from live neutrophils occurs independent of RIPK3 and MLKL (5).

Phagocytosis of methicillin-resistant Staphylococcus aureus can also induce necroptosis in human neutrophils, as some ingested bacteria may survive within the phagosome and prevent phagocytosis-induced apoptosis (94, 95). This requires RIPK3 but is independent of RIPK1 and MLKL (94) and is associated with neutrophil serine protease-mediated IL-1β secretion (141, 278). The mechanism of S. aureus-evoked IL-1β production in neutrophils differs from that in macrophages, in which it is mediated by the canonical NLRP3 inflammasome and caspase-1 (278). Necroptosis may allow the escape of viable S. aureus from dead PMNs, liberation of intracellular neutrophil contents, and generation of IL-1β, which aggravate local tissue injury and lead to persistent infection (95). Activation of the RIPK3-MLKL pathway was detected in neutrophils in inflamed tissue samples from patients with neutrophilic diseases (289), indicating that neutrophil necroptosis occurs under in vivo inflammatory conditions, although the relevance of necroptosis to these pathologies remains elusive.

Dying cells release a myriad of DAMPs, including mitochondrial formyl peptides, purines, LTB4, cytokines, and chemokines, and necrotic debris activates the complement cascade to generate the chemoattractant C3a and C5a, which collectively serve as necrotic “find me” signals (38, 296). These signals guide recruitment of phagocytes to the vicinity of necrotic targets (184, 296). Necrotic cells also express “eat me” ligands that facilitate their uptake. Some of these ligands partially overlap with equivalent apoptotic signals (e.g., exposure of phosphatidylserine and LTB4) (296), whereas others, such as complement C1q deposited on necrotic cell membrane (85) and externalization of annexin A1 on the surface of necrotic cells (23), are unique to necrotic debris. Little is known about the ligands and receptors mediating the clearance of necrotic neutrophils. The importance of removing necrotic PMNs is illustrated by the detrimental effect of neutrophil antigens such as proteinase-3 to trigger autoimmunity, as reported in vasculitis (113, 232).

Therapeutic Approaches to Facilitate Neutrophil Clearance from Inflamed Tissues

Resolution of inflammation is an active process enabling the affected tissues to restore function (253, 257). The resolution phase of self-limiting inflammation is skewed toward proresolving mediators, as illustrated by changes in lipid profile with increases in generation of LXA4 and resolvin D5 in a mouse model of bacterial peritonitis (39). Conversely, insufficient activation of resolution mechanisms contributes to excessive or prolonged inflammatory response to the initial insult (197, 253). For example, stable atherosclerotic plaques exhibit a controlled chronic inflammation phenotype, whereas defect in proresolution mechanisms precedes plaque rupture (77, 78). Ligation of TLR9 is associated with markedly reduced production of proresolving lipid mediators, such as 15-epi-LXA4 and RvD1, which likely contributes to propagation of bacterial infections in mice (250). Thus, restoring dysregulated or defective endogenous resolution circuits is anticipated to promote resolution by correcting the imbalance between proinflammatory and proresolving mediators. Indeed, recent research identified many resolution mediators as promising agents to target neutrophils to achieve this goal. Table 1 summarizes results obtained in animal disease models and patients.

Table 1.

Potential therapeutic approaches to target neutrophils for promoting resolution of inflammation

Molecule Receptor Species Disease/Model Key Mechanisms, Actions on Neutrophils Effects on Disease Reference
Annexin A1 ALX/FPR2 Mouse Mesenteric circulation Detachment of adherent PMNs, ↓emigration 215
Mouse Dermatitis ↓Inflammation 160
Mouse Zymosan-induced dermatitis ↑monocyte recruitment, ↑efferocytosis ↓Inflammation 183
Mouse Blood-neutrophil senescence ↓PMN senescence, ↑migratory capacity ↑PMN clearance in bone marrow 52, 167
Peptide Ac 2-26 FPR1, ALX/FPR2 Rat Peritonitis ↑macrophage accumulation, ↓PMN accumulation ↓Inflammation
↓Systemic inflammation
266
Mouse Intestinal ischemia-reperfusion-induced lung injury ↑IL-10, ↓PMN recruitment into the lung ↓Lung injury 96
Mouse Pleurisy ↓Mcl-1 expression, ↓NF-κB, ↓ERK 1/2, ↑caspase-3, ↑PMN apoptosis ↑Resolution 283
Mouse Skin allograft ↓Transmigration, ↑PMN apoptosis ↓Tissue damage, ↑graft survival 274
LXA4, 15-epi-LXA4 ALX/FPR2 Mouse Asthma ↓Prostanoids, ↓IL-5, ↓IL-13, ↓cysteinyl LTs ↓Airway inflammation, ↓hypersensitivity 156
Mouse Ischemia-reperfusion injury-induced lung injury ↓BLT signaling, ↓5-LO signaling, ↓PMN accumulation ↓Lung injury 40
Mouse Peritonitis ↓PMN accumulation, ↑phagocytosis, ↑efferocytosis, ↑PMN migration to lymph nodes, ↑IL-10, ↑efferocytosis ↑Resolution 92, 248
Mouse Cystic fibrosis ↓PMN accumulation ↓Bacterial burden, ↓disease severity 126
Mouse ARDS, acute lung injury Restores impaired phagocytosis, ↓NE and PR3 release, ↑PMN apoptosis, ↑efferocytosis ↑Bacterial clearance, ↑resolution 250
Mouse ARDS, acute lung injury ↓MPO signaling, ↓Mcl-1, ↑PMN apoptosis, ↑efferocytosis ↑Resolution, ↑survival 69
Rabbit Periodontitis ↓LTB4 signaling, ↓PMN accumulation ↓Inflammation, ↓bone loss 256
Resolvin D1 GPR32 Mouse Peritonitis ↓PMN transendothelial migration, ↓PMN accumulation, ↑bacterial clearance, ↑IL-10, ↑phagocytosis, ↑efferocytosis ↓Inflammation 247, 268
Mouse Renal ischemia-reperfusion injury ↓TLR-mediated activation of macrophages, ↓PMN infiltration ↑Renal function, ↓fibrosis 59
17-epi-
resolvin D1
ALX/FPR2 Mouse ARDS, acute lung injury Restores impaired phagocytosis, ↓NE and PR3 release, ↑bacterial killing, ↑PMN apoptosis, ↑efferocytosis ↑Bacterial clearance ↑Resolution 250
Resolvin D2 Mouse Microbial sepsis, peritonitis ↓PMN accumulation, ↑bacterial clearance, ↑peritoneal macrophage ↓Inflammation, ↑survival 264
Resolvin D5 GPR32 Mouse Bacterial peritonitis ↓PMN infiltration, ↑PMN phagocytosis, ↑bacterial clearance ↓Antibiotic need, ↑resolution, ↑survival 39
Resolvin E1 ChemR23 Mouse Peritonitis ↓PMN trafficking, ↑efferocytosis, ↑lymphatic PMN, ↓IL-12, ↓NF-κB ↓Inflammation 10, 247, 248
Mouse Colitis ↓PMN recruitment ↓Inflammation, ↑survival 12
BLT1, ChemR23 Mouse Peritonitis Partial BLT1 agonist/antagonist, ↓LTB4 signaling, ↓PMN trafficking, ↓IL-12, ↓TNF-α, ↓iNOS ↓Inflammation, ↓body weight loss, ↑survival 10, 11, 248
Mouse Bacterial pneumonia ↓PMN recruitment, ↑bacterial clearance, ↓IL-1β, ↓HMGB1 ↓Inflammation, ↑survival 251
BLT2 Mouse Acute lung injury, peritonitis-associated lung injury ↓PMN accumulation, ↑phagocytosis, ↑PMN apoptosis, ↑efferocytosis ↓Tissue injury, ↑resolution
↑survival
66
Rabbit Periodontitis ↓PMN infiltration ↓Inflammation, ↓bone loss, 106
Maresin 1 ? Mouse Peritonitis ↓PMN accumulation, ↑efferocytosis ↓inflammation 259
Mouse ARDS ↑Platelet-PMN interactions, ↓PMN trafficking, ↑efferocytosis, Organ protection, ↓tissue hypoxia, ↓edema 2
Protectin D1 ? Mouse Renal ischemia-reperfusion injury ↓PMN infiltration, ↓TLR-mediated macrophage activation ↑Renal function, ↓fibrosis 59
Mouse Peritonitis ↓PMN infiltration, ↑phagocytosis, ↑efferocytosis, ↑lymphatic PMN ↑Resolution 248, 255
Galectin 1 Mouse Peritonitis ↓PMN adherence, ↑PMN clearance, ↑IFN-β, ↓PR3 ↑Resolution 150, 301
Protein S/GAS6 TAM receptors Mouse Periodontitis ↓PMN accumulation,↑ bacterial clearance, ↑IL-10 ↑Resolution 196
Mouse Acute lung injury ↑PMN clearance, ↑efferocytosis ↑Resolution 200
DEL-1 LFA1 (CD11a) Mouse Acute lung injury ↓PMN accumulation ↑Resolution 42
Mouse Periodontitis ↓PMN accumulation,↓TNF-α, ↓IL-17 ↑Resolution
↓Bone loss
71
Phosphatidylserine,
αVβ3 integrin
Mouse Peritonitis ↓PMN infiltration, ↑RvD1, ↑RvE1, ↑efferocytosis 139
Interferon-β IFNα/βR1 Mouse Bacterial pneumonia, peritonitis ↑PMN apoptosis, ↑bacterial clearance, ↑efferocytosis, ↓Mcl-1, ↑IL-10,↓IL-6, ↓TNF-α ↑Resolution 242
Elafin (PI3) NE inhibitor Human Early phase ARDS ↓NE activity ↓Plasma NE level biomarker? 292
Mouse Pleurisy Mobilization of annexin A1, ↓NE activity, ↑PMN apoptosis,↑M2 macrophages, ↑efferocytosis ↑Resolution 284
Mouse Gout Similar actions as in pleurisy ↓Inflammation 284
PF-1355 MPO inhibitor Mouse Immune complex vasculitis ↓MPO activity, ↓PMN recruitment, ↓NET formation ↓Pulmonary vascular injury 310
Mouse Anti-GBM disease ↓MPO activity, ↓PMN recruitment, ↓NET formation ↑Renal function 310
Anti-TNF antibody TNF neutralization Human Rheumatoid arthritis ↓NET formation ex vivo, ↓ expression of PMN auto-antigens ? 131
Agonistic FAS antibody FAS Human Severe trauma Stimulation of FAS, ↓Mcl-1, ↑PMN apoptosis ex vivo ? 214
R-roscovitine, NG 75 Inhibition of cyclin-dependent kinases Mouse Pleurisy, Aacute lung injury ↓PMN accumulation, ↑apoptosis, ↑efferocytosis ↑Resolution 238
Human Cystic fibrosis Restores suppressed PMN apoptosis ex vivo ? 189
Hydrogen sulfide Mouse Endotoxemia in the mesenteric circulation ↑annexin A1 mobilization, ↓PMN trafficking ↓Inflammation 27
Nedocromil Mast cell stabilization Mouse Mesenteric ischemia-reperfusion injury ↑annexin A1 mobilization, ↓PMN accumulation ↓Tissue injury 302
DNase I Mouse ARDS ↑NET degradation ↓Lung injury 153, 158
Mouse Systemic lupus erythematosus ↑NET degradation ↓Autoantibodies, ↓mortality 166
Dornase-α (synthetic DNase I) Human Respiratory failure after severe trauma ↑NET degradation ↓Respiratory failure 229

ARDS, acute respiratory distress syndrome; BLT1, leukotriene B4 receptor 1; BLT2, leukotriene B4 receptor 2; DEL-1, developmental endothelial locus-1; iNOS, inducible NO synthase; 5-LO, 5-lipoxygenase; LT, leukotriene; MPO, myeloperoxidase; NE, neutrophil elastase; NET, neutrophil extracellular trap; PI3, phosphoinositide 3; PMN, polymorphonuclear leukocytes; PR3, proteinase-3; TLR, toll-like receptor; ? indicates yet undefined receptor/effect.

Stopping PMN recruitment into inflamed sites and stimulating efferocytosis are common features of most proresolving lipid (253, 257) and protein mediators (150, 218). These mediators possess multipronged, somewhat overlapping functions but also express special cell-specific functions mediated through distinct receptors and mechanisms in the resolution phase. ALX/FPR2 binds multiple proresolving ligands, including annexin A1, annexin A1-derived peptide Ac2-26 (217), LXA4 and its aspirin-triggered 15-epimeric form 15-epi-LXA4 (253, 257), and 17-epi-RvD1 (257), whereas RvD1 and RvD5 signals through GPR32 (39) and RvE1 signals through ChemR23 (268) and BLT1 (66, 253). Ligation of these receptors was found to reduce tissue injury (10, 12, 35, 69, 96, 126, 250, 274), decrease bacterial burden (39, 126, 250), and enhance phagocytosis (39, 66, 92, 247, 248, 250, 251, 255, 256, 264), PMN apoptosis (69, 70, 250, 274, 283), and efferocytosis (2, 10, 69, 70, 92, 167, 183, 247, 248, 250, 255, 259, 268), contributing to preservation of organ functions (2, 59, 106, 156), acceleration of resolution (2, 1012, 39, 40, 42, 52, 59, 69, 71, 92, 96, 106, 126, 131, 156, 160, 167, 183, 189, 196, 200, 214, 247, 248, 250, 251, 255, 256, 259, 264, 266, 268, 283, 292, 300, 310), and increased survival (39, 69, 248, 251, 264) under a variety of pathological conditions (Table 1). There is evidence that some of these mediators form feedforward amplifying loops, for example, LXA4-annexin A1 (26) and RvE1-LXA4 (107), to counter PMN recruitment and activation, thereby driving resolution. ALX/FPR2 agonists can be harnessed in conjunction with antibiotics (39) or antiviral drugs (192) to fortify the host response to infections (204, 219).

Pharmacological induction of PMN apoptosis with cyclin-dependent kinase inhibitors (238) or synthetic annexin A1-derived peptides such as peptide Ac 2-26 (274, 283) mimic the resolution enhancing actions of SPMs. The clinical potential of this approach is illustrated by the efficacy of an agonistic FAS antibody to counter suppressed apoptosis in PMN from patients with severe trauma ex vivo (214). The gaseous mediator H2S (27) and the mast cell-stabilizing drug nedocromil (302) were shown to counter the deleterious actions of PMN through mobilization of annexin A1. Pharmacological blockade of the activity of PMN-derived enzymes, neutrophil elastase, or myeloperoxidase attenuated tissue injury and promoted resolution in murine pleurisy and immune complex vasculitis (284, 310). The neutrophil elastase inhibitor elafin efficiently reduced plasma elastase activity in patients with ARDS (292), suggesting the potential use of elafin as a biomarker of the disease. Reducing NET formation or accelerating its degradation with DNase I (153, 158), MPO inhibitor (310), and anti-TNF antibody (214) is under investigations to prevent propagation of organ damage and autoimmune responses (98). Of note is that the synthetic DNase I analog dornase-α is currently being tested in phase III clinical trials in patients with severe trauma-associated respiratory failure (229). Because presentation of severe COVID-19 resembles known NET-associated pathologies ARDS and microthrombosis (17, 312), targeting neutrophils or NET formation may represent promising approaches in the treatment for COVID-19.

Sex differences in the resolution of inflammation have been observed in mice (216, 288) and humans (234). Studies on skin blisters in healthy volunteers suggest accelerated resolution in females compared with males, presumably due to reduced PMN accumulation and elevated levels of D-series resolvins (234). Future studies are warranted to further explore differences in resolution mechanisms between sexes.

CONCLUDING REMARKS

Consideration of neutrophil functional heterogeneity and novel paradigms in neutrophil biology provides a different vantage point of their roles in host defense and disease. Clearance of neutrophils from infected or injured tissues is critical for efficient resolution and return to homeostasis. Impaired PMN removal has harmful consequences. Therefore, it is essential to further explore the mechanisms that determine the fate of emigrated neutrophils. These mechanisms govern phenotypical and functional heterogeneity of neutrophils, although linking phenotype to specific functions is challenging. Understanding the diversity in PMN functions is essential to develop neutrophil-specific therapies. The current anti-inflammatory drugs alleviate symptoms of inflammation (swelling, pain) but do not promote and may even delay resolution and tissue repair. An attractive alternative approach would be to stimulate egress of neutrophils from inflammatory loci. Results from preclinical models indicate that this can be accomplished by the application of SPMs such as lipoxins, resolvins, annexin A1, galectin 1, protein S, and IFN-β, which can prevent the deleterious effects of excessive or prolonged PMN-mediated inflammation. Although large-scale clinical trials with these compounds seem distant, initial results with targeting neutrophil-specific functions (e.g., inhibition of neutrophil elastase or degradation of NET) hold promise as disease-modifying intervention. Further studies are needed to investigate whether therapeutic interventions to facilitate removal of pathogenic neutrophils from inflamed tissues will lead to resolution of inflammation underlying many chronic diseases.

GRANTS

This work was supported by Canadian Institutes of Health Research Grants MOP-97742 and MOP-102619 (to J.G.F.), Israel Science Foundation Grant No. 678/13, the Rosetrees Trust, and the Wolfson Family Charitable Trust (to A.A.).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

J.G.F. and A.A. prepared figures; J.G.F. and A.A. drafted manuscript; J.G.F. and A.A. edited and revised manuscript; J.G.F. and A.A. approved final version of manuscript.

REFERENCES

  • 1.Abadie V, Badell E, Douillard P, Ensergueix D, Leenen PJ, Tanguy M, Fiette L, Saeland S, Gicquel B, Winter N. Neutrophils rapidly migrate via lymphatics after Mycobacterium bovis BCG intradermal vaccination and shuttle live bacilli to the draining lymph nodes. Blood 106: 1843–1850, 2005. doi: 10.1182/blood-2005-03-1281. [DOI] [PubMed] [Google Scholar]
  • 2.Abdulnour RE, Dalli J, Colby JK, Krishnamoorthy N, Timmons JY, Tan SH, Colas RA, Petasis NA, Serhan CN, Levy BD. Maresin 1 biosynthesis during platelet-neutrophil interactions is organ-protective. Proc Natl Acad Sci USA 111: 16526–16531, 2014. doi: 10.1073/pnas.1407123111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Adrover JM, Del Fresno C, Crainiciuc G, Cuartero MI, Casanova-Acebes M, Weiss LA, Huerga-Encabo H, Silvestre-Roig C, Rossaint J, Cossío I, Lechuga-Vieco AV, García-Prieto J, Gómez-Parrizas M, Quintana JA, Ballesteros I, Martin-Salamanca S, Aroca-Crevillen A, Chong SZ, Evrard M, Balabanian K, López J, Bidzhekov K, Bachelerie F, Abad-Santos F, Muñoz-Calleja C, Zarbock A, Soehnlein O, Weber C, Ng LG, Lopez-Rodriguez C, Sancho D, Moro MA, Ibáñez B, Hidalgo A. A neutrophil timer coordinates immune defense and vascular protection. Immunity 50: 390–402.e10, 2019. [Erratum in Immunity 51: 966–967, 2019]. doi: 10.1016/j.immuni.2019.01.002. [DOI] [PubMed] [Google Scholar]
  • 4.Afonso PV, Janka-Junttila M, Lee YJ, McCann CP, Oliver CM, Aamer KA, Losert W, Cicerone MT, Parent CA. LTB4 is a signal-relay molecule during neutrophil chemotaxis. Dev Cell 22: 1079–1091, 2012. doi: 10.1016/j.devcel.2012.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Amini P, Stojkov D, Wang X, Wicki S, Kaufmann T, Wong WW, Simon HU, Yousefi S. NET formation can occur independently of RIPK3 and MLKL signaling. Eur J Immunol 46: 178–184, 2016. doi: 10.1002/eji.201545615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Köckritz-Blickwede M, Schilling B, Brandau S, Weiss S, Jablonska J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer 138: 1982–1993, 2016. doi: 10.1002/ijc.29945. [DOI] [PubMed] [Google Scholar]
  • 7.Andzinski L, Wu CF, Lienenklaus S, Kröger A, Weiss S, Jablonska J. Delayed apoptosis of tumor associated neutrophils in the absence of endogenous IFN-β. Int J Cancer 136: 572–583, 2015. doi: 10.1002/ijc.28957. [DOI] [PubMed] [Google Scholar]
  • 8.Ariel A, Fredman G, Sun YP, Kantarci A, Van Dyke TE, Luster AD, Serhan CN. Apoptotic neutrophils and T cells sequester chemokines during immune response resolution through modulation of CCR5 expression. Nat Immunol 7: 1209–1216, 2006. doi: 10.1038/ni1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ariel A, Serhan CN. New lives given by cell death: macrophage differentiation following their encounter with apoptotic leukocytes during the resolution of inflammation. Front Immunol 3: 4, 2012. doi: 10.3389/fimmu.2012.00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Arita M, Bianchini F, Aliberti J, Sher A, Chiang N, Hong S, Yang R, Petasis NA, Serhan CN. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J Exp Med 201: 713–722, 2005. doi: 10.1084/jem.20042031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Arita M, Ohira T, Sun YP, Elangovan S, Chiang N, Serhan CN. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. J Immunol 178: 3912–3917, 2007. doi: 10.4049/jimmunol.178.6.3912. [DOI] [PubMed] [Google Scholar]
  • 12.Arita M, Yoshida M, Hong S, Tjonahen E, Glickman JN, Petasis NA, Blumberg RS, Serhan CN. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proc Natl Acad Sci USA 102: 7671–7676, 2005. doi: 10.1073/pnas.0409271102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Aswad M, Assi S, Schif-Zuck S, Ariel A. CCL5 promotes resolution-phase macrophage reprogramming in concert with the atypical chemokine receptor D6 and apoptotic polymorphonuclear cells. J Immunol 199: 1393–1404, 2017. doi: 10.4049/jimmunol.1502542. [DOI] [PubMed] [Google Scholar]
  • 14.Balza E, Piccioli P, Carta S, Lavieri R, Gattorno M, Semino C, Castellani P, Rubartelli A. Proton pump inhibitors protect mice from acute systemic inflammation and induce long-term cross-tolerance. Cell Death Dis 7: e2304, 2016. doi: 10.1038/cddis.2016.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Banerjee S, de Freitas A, Friggeri A, Zmijewski JW, Liu G, Abraham E. Intracellular HMGB1 negatively regulates efferocytosis. J Immunol 187: 4686–4694, 2011. doi: 10.4049/jimmunol.1101500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bardoel BW, Kenny EF, Sollberger G, Zychlinsky A. The balancing act of neutrophils. Cell Host Microbe 15: 526–536, 2014. doi: 10.1016/j.chom.2014.04.011. [DOI] [PubMed] [Google Scholar]
  • 17.Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, Daßler-Plenker J, Guerci P, Huynh C, Knight JS, Loda M, Looney MR, McAllister F, Rayes R, Renaud S, Rousseau S, Salvatore S, Schwartz RE, Spicer JD, Yost CC, Weber A, Zuo Y, Egeblad M. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J Exp Med 217: e20200652, 2020. doi: 10.1084/jem.20200652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bauer S, Abdgawad M, Gunnarsson L, Segelmark M, Tapper H, Hellmark T. Proteinase 3 and CD177 are expressed on the plasma membrane of the same subset of neutrophils. J Leukoc Biol 81: 458–464, 2007. doi: 10.1189/jlb.0806514. [DOI] [PubMed] [Google Scholar]
  • 19.Benarafa C, Simon HU. Role of granule proteases in the life and death of neutrophils. Biochem Biophys Res Commun 482: 473–481, 2017. doi: 10.1016/j.bbrc.2016.11.086. [DOI] [PubMed] [Google Scholar]
  • 20.Berger M, Sorensen RU, Tosi MF, Dearborn DG, Döring G. Complement receptor expression on neutrophils at an inflammatory site, the Pseudomonas-infected lung in cystic fibrosis. J Clin Invest 84: 1302–1313, 1989. doi: 10.1172/JCI114298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Beyrau M, Bodkin JV, Nourshargh S. Neutrophil heterogeneity in health and disease: a revitalized avenue in inflammation and immunity. Open Biol 2: 120134, 2012. doi: 10.1098/rsob.120134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol 81: 1–5, 2007. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 23.Blume KE, Soeroes S, Waibel M, Keppeler H, Wesselborg S, Herrmann M, Schulze-Osthoff K, Lauber K. Cell surface externalization of annexin A1 as a failsafe mechanism preventing inflammatory responses during secondary necrosis. J Immunol 183: 8138–8147, 2009. doi: 10.4049/jimmunol.0902250. [DOI] [PubMed] [Google Scholar]
  • 24.Bozinovski S, Uddin M, Vlahos R, Thompson M, McQualter JL, Merritt AS, Wark PA, Hutchinson A, Irving LB, Levy BD, Anderson GP. Serum amyloid A opposes lipoxin A4 to mediate glucocorticoid refractory lung inflammation in chronic obstructive pulmonary disease. Proc Natl Acad Sci USA 109: 935–940, 2012. doi: 10.1073/pnas.1109382109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Brackett CM, Muhitch JB, Evans SS, Gollnick SO. IL-17 promotes neutrophil entry into tumor-draining lymph nodes following induction of sterile inflammation. J Immunol 191: 4348–4357, 2013. doi: 10.4049/jimmunol.1103621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Brancaleone V, Dalli J, Bena S, Flower RJ, Cirino G, Perretti M. Evidence for an anti-inflammatory loop centered on polymorphonuclear leukocyte formyl peptide receptor 2/lipoxin A4 receptor and operative in the inflamed microvasculature. J Immunol 186: 4905–4914, 2011. doi: 10.4049/jimmunol.1003145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Brancaleone V, Mitidieri E, Flower RJ, Cirino G, Perretti M. Annexin A1 mediates hydrogen sulfide properties in the control of inflammation. J Pharmacol Exp Ther 351: 96–104, 2014. doi: 10.1124/jpet.114.217034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science 303: 1532–1535, 2004. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
  • 29.Brovkovych V, Gao XP, Ong E, Brovkovych S, Brennan ML, Su X, Hazen SL, Malik AB, Skidgel RA. Augmented inducible nitric oxide synthase expression and increased NO production reduce sepsis-induced lung injury and mortality in myeloperoxidase-null mice. Am J Physiol Lung Cell Mol Physiol 295: L96–L103, 2008. doi: 10.1152/ajplung.00450.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Buckley CD, Ross EA, McGettrick HM, Osborne CE, Haworth O, Schmutz C, Stone PC, Salmon M, Matharu NM, Vohra RK, Nash GB, Rainger GE. Identification of a phenotypically and functionally distinct population of long-lived neutrophils in a model of reverse endothelial migration. J Leukoc Biol 79: 303–311, 2006. doi: 10.1189/jlb.0905496. [DOI] [PubMed] [Google Scholar]
  • 31.Butenko S, Satyanarayanan SK, Assi S, Schif-Zuck S, Barkan D, Sher N, Ariel A. Transcriptomic analysis of monocyte-derived non-phagocytic macrophages favors a role in limiting tissue repair and fibrosis. Front Immunol 11: 405, 2020. [Erratum in Front Immunol 11: 1003, 2020]. doi: 10.3389/fimmu.2020.00405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cai TQ, Wright SD. Human leukocyte elastase is an endogenous ligand for the integrin CR3 (CD11b/CD18, Mac-1, alpha M beta 2) and modulates polymorphonuclear leukocyte adhesion. J Exp Med 184: 1213–1223, 1996. doi: 10.1084/jem.184.4.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Campana L, Starkey Lewis PJ, Pellicoro A, Aucott RL, Man J, O’Duibhir E, Mok SE, Ferreira-Gonzalez S, Livingstone E, Greenhalgh SN, Hull KL, Kendall TJ, Vernimmen D, Henderson NC, Boulter L, Gregory CD, Feng Y, Anderton SM, Forbes SJ, Iredale JP. The STAT3-IL-10-IL-6 pathway is a novel regulator of macrophage efferocytosis and phenotypic conversion in sterile liver injury. J Immunol 200: 1169–1187, 2018. doi: 10.4049/jimmunol.1701247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Carmona-Rivera C, Kaplan MJ. Low-density granulocytes: a distinct class of neutrophils in systemic autoimmunity. Semin Immunopathol 35: 455–463, 2013. doi: 10.1007/s00281-013-0375-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Casanova-Acebes M, Pitaval C, Weiss LA, Nombela-Arrieta C, Chèvre R, A-González N, Kunisaki Y, Zhang D, van Rooijen N, Silberstein LE, Weber C, Nagasawa T, Frenette PS, Castrillo A, Hidalgo A. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 153: 1025–1035, 2013. doi: 10.1016/j.cell.2013.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chatterjee BE, Yona S, Rosignoli G, Young RE, Nourshargh S, Flower RJ, Perretti M. Annexin 1-deficient neutrophils exhibit enhanced transmigration in vivo and increased responsiveness in vitro. J Leukoc Biol 78: 639–646, 2005. doi: 10.1189/jlb.0405206. [DOI] [PubMed] [Google Scholar]
  • 37.Chen F, Wu W, Millman A, Craft JF, Chen E, Patel N, Boucher JL, Urban JF Jr, Kim CC, Gause WC. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat Immunol 15: 938–946, 2014. doi: 10.1038/ni.2984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 10: 826–837, 2010. doi: 10.1038/nri2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chiang N, Fredman G, Bäckhed F, Oh SF, Vickery T, Schmidt BA, Serhan CN. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 484: 524–528, 2012. doi: 10.1038/nature11042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chiang N, Gronert K, Clish CB, O’Brien JA, Freeman MW, Serhan CN. Leukotriene B4 receptor transgenic mice reveal novel protective roles for lipoxins and aspirin-triggered lipoxins in reperfusion. J Clin Invest 104: 309–316, 1999. doi: 10.1172/JCI7016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chiang N, Libreros S, Norris PC, de la Rosa X, Serhan CN. Maresin 1 activates LGR6 receptor promoting phagocyte immunoresolvent functions. J Clin Invest 129: 5294–5311, 2019. doi: 10.1172/JCI129448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Choi EY, Chavakis E, Czabanka MA, Langer HF, Fraemohs L, Economopoulou M, Kundu RK, Orlandi A, Zheng YY, Prieto DA, Ballantyne CM, Constant SL, Aird WC, Papayannopoulou T, Gahmberg CG, Udey MC, Vajkoczy P, Quertermous T, Dimmeler S, Weber C, Chavakis T. Del-1, an endogenous leukocyte-endothelial adhesion inhibitor, limits inflammatory cell recruitment. Science 322: 1101–1104, 2008. doi: 10.1126/science.1165218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chtanova T, Schaeffer M, Han SJ, van Dooren GG, Nollmann M, Herzmark P, Chan SW, Satija H, Camfield K, Aaron H, Striepen B, Robey EA. Dynamics of neutrophil migration in lymph nodes during infection. Immunity 29: 487–496, 2008. doi: 10.1016/j.immuni.2008.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 13: 463–469, 2007. doi: 10.1038/nm1565. [DOI] [PubMed] [Google Scholar]
  • 45.Clemmensen SN, Bohr CT, Rørvig S, Glenthøj A, Mora-Jensen H, Cramer EP, Jacobsen LC, Larsen MT, Cowland JB, Tanassi JT, Heegaard NH, Wren JD, Silahtaroglu AN, Borregaard N. Olfactomedin 4 defines a subset of human neutrophils. J Leukoc Biol 91: 495–500, 2012. doi: 10.1189/jlb.0811417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Colom B, Bodkin JV, Beyrau M, Woodfin A, Ody C, Rourke C, Chavakis T, Brohi K, Imhof BA, Nourshargh S. Leukotriene B4-neutrophil elastase axis drives neutrophil reverse transendothelial cell migration in vivo. Immunity 42: 1075–1086, 2015. doi: 10.1016/j.immuni.2015.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Colotta F, Re F, Polentarutti N, Sozzani S, Mantovani A. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood 80: 2012–2020, 1992. doi: 10.1182/blood.V80.8.2012.2012. [DOI] [PubMed] [Google Scholar]
  • 48.Coombes JL, Charsar BA, Han SJ, Halkias J, Chan SW, Koshy AA, Striepen B, Robey EA. Motile invaded neutrophils in the small intestine of Toxoplasma gondii-infected mice reveal a potential mechanism for parasite spread. Proc Natl Acad Sci USA 110: E1913–E1922, 2013. doi: 10.1073/pnas.1220272110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Coombs C, Georgantzoglou A, Walker HA, Patt J, Merten N, Poplimont H, Busch-Nentwich EM, Williams S, Kotsi C, Kostenis E, Sarris M. Chemokine receptor trafficking coordinates neutrophil clustering and dispersal at wounds in zebrafish. Nat Commun 10: 5166, 2019. [Erratum in Nat Commun 11: 506, 2020]. doi: 10.1038/s41467-019-13107-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cooray SN, Gobbetti T, Montero-Melendez T, McArthur S, Thompson D, Clark AJL, Flower RJ, Perretti M. Ligand-specific conformational change of the G-protein-coupled receptor ALX/FPR2 determines proresolving functional responses. Proc Natl Acad Sci USA 110: 18232–18237, 2013. doi: 10.1073/pnas.1308253110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dalli J, Consalvo AP, Ray V, Di Filippo C, D’Amico M, Mehta N, Perretti M. Proresolving and tissue-protective actions of annexin A1-based cleavage-resistant peptides are mediated by formyl peptide receptor 2/lipoxin A4 receptor. J Immunol 190: 6478–6487, 2013. doi: 10.4049/jimmunol.1203000. [DOI] [PubMed] [Google Scholar]
  • 52.Dalli J, Jones CP, Cavalcanti DM, Farsky SH, Perretti M, Rankin SM. Annexin A1 regulates neutrophil clearance by macrophages in the mouse bone marrow. FASEB J 26: 387–396, 2012. doi: 10.1096/fj.11-182089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, McCune WJ, Kaplan MJ. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 184: 3284–3297, 2010. doi: 10.4049/jimmunol.0902199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Desai J, Kumar SV, Mulay SR, Konrad L, Romoli S, Schauer C, Herrmann M, Bilyy R, Müller S, Popper B, Nakazawa D, Weidenbusch M, Thomasova D, Krautwald S, Linkermann A, Anders HJ. PMA and crystal-induced neutrophil extracellular trap formation involves RIPK1-RIPK3-MLKL signaling. Eur J Immunol 46: 223–229, 2016. doi: 10.1002/eji.201545605. [DOI] [PubMed] [Google Scholar]
  • 55.Desai J, Mulay SR, Nakazawa D, Anders HJ. Matters of life and death. How neutrophils die or survive along NET release and is “NETosis” = necroptosis? Cell Mol Life Sci 73: 2211–2219, 2016. doi: 10.1007/s00018-016-2195-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dieker J, Tel J, Pieterse E, Thielen A, Rother N, Bakker M, Fransen J, Dijkman HB, Berden JH, de Vries JM, Hilbrands LB, van der Vlag J. Circulating apoptotic microparticles in systemic lupus erythematosus patients drive the activation of dendritic cell subsets and prime neutrophils for NETosis. Arthritis Rheumatol 68: 462–472, 2016. doi: 10.1002/art.39417. [DOI] [PubMed] [Google Scholar]
  • 57.Doran AC, Yurdagul A Jr, Tabas I. Efferocytosis in health and disease. Nat Rev Immunol 20: 254–267, 2020. doi: 10.1038/s41577-019-0240-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Douda DN, Khan MA, Grasemann H, Palaniyar N. SK3 channel and mitochondrial ROS mediate NADPH oxidase-independent NETosis induced by calcium influx. Proc Natl Acad Sci USA 112: 2817–2822, 2015. doi: 10.1073/pnas.1414055112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Duffield JS, Hong S, Vaidya VS, Lu Y, Fredman G, Serhan CN, Bonventre JV. Resolvin D series and protectin D1 mitigate acute kidney injury. J Immunol 177: 5902–5911, 2006. doi: 10.4049/jimmunol.177.9.5902. [DOI] [PubMed] [Google Scholar]
  • 60.Duffy D, Perrin H, Abadie V, Benhabiles N, Boissonnas A, Liard C, Descours B, Reboulleau D, Bonduelle O, Verrier B, Van Rooijen N, Combadière C, Combadière B. Neutrophils transport antigen from the dermis to the bone marrow, initiating a source of memory CD8+ T cells. Immunity 37: 917–929, 2012. doi: 10.1016/j.immuni.2012.07.015. [DOI] [PubMed] [Google Scholar]
  • 61.Dupuy AG, Caron E. Integrin-dependent phagocytosis: spreading from microadhesion to new concepts. J Cell Sci 121: 1773–1783, 2008. doi: 10.1242/jcs.018036. [DOI] [PubMed] [Google Scholar]
  • 62.Dzhagalov I, St. John A, He YW. The antiapoptotic protein Mcl-1 is essential for the survival of neutrophils but not macrophages. Blood 109: 1620–1626, 2007. doi: 10.1182/blood-2006-03-013771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Eash KJ, Greenbaum AM, Gopalan PK, Link DC. CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow. J Clin Invest 120: 2423–2431, 2010. doi: 10.1172/JCI41649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Edlow DW, Sheldon WH. The pH of inflammatory exudates. Proc Soc Exp Biol Med 137: 1328–1332, 1971. doi: 10.3181/00379727-137-35782. [DOI] [PubMed] [Google Scholar]
  • 65.El Kebir D, de Oliveira Lima Dos Santos E, Mansouri S, Sekheri M, Filep JG. Mild acidosis delays neutrophil apoptosis via multiple signaling pathways and acts in concert with inflammatory mediators. J Leukoc Biol 102: 1389–1400, 2017. doi: 10.1189/jlb.3A0117-041R. [DOI] [PubMed] [Google Scholar]
  • 66.El Kebir D, Gjorstrup P, Filep JG. Resolvin E1 promotes phagocytosis-induced neutrophil apoptosis and accelerates resolution of pulmonary inflammation. Proc Natl Acad Sci USA 109: 14983–14988, 2012. doi: 10.1073/pnas.1206641109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.El Kebir D, József L, Khreiss T, Pan W, Petasis NA, Serhan CN, Filep JG. Aspirin-triggered lipoxins override the apoptosis-delaying action of serum amyloid A in human neutrophils: a novel mechanism for resolution of inflammation. J Immunol 179: 616–622, 2007. doi: 10.4049/jimmunol.179.1.616. [DOI] [PubMed] [Google Scholar]
  • 68.El Kebir D, József L, Pan W, Filep JG. Myeloperoxidase delays neutrophil apoptosis through CD11b/CD18 integrins and prolongs inflammation. Circ Res 103: 352–359, 2008. doi: 10.1161/01.RES.0000326772.76822.7a. [DOI] [PubMed] [Google Scholar]
  • 69.El Kebir D, József L, Pan W, Wang L, Petasis NA, Serhan CN, Filep JG. 15-epi-lipoxin A4 inhibits myeloperoxidase signaling and enhances resolution of acute lung injury. Am J Respir Crit Care Med 180: 311–319, 2009. doi: 10.1164/rccm.200810-1601OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Erreni M, Manfredi AA, Garlanda C, Mantovani A, Rovere-Querini P. The long pentraxin PTX3: A prototypical sensor of tissue injury and a regulator of homeostasis. Immunol Rev 280: 112–125, 2017. doi: 10.1111/imr.12570. [DOI] [PubMed] [Google Scholar]
  • 71.Eskan MA, Jotwani R, Abe T, Chmelar J, Lim JH, Liang S, Ciero PA, Krauss JL, Li F, Rauner M, Hofbauer LC, Choi EY, Chung KJ, Hashim A, Curtis MA, Chavakis T, Hajishengallis G. The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat Immunol 13: 465–473, 2012. doi: 10.1038/ni.2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Evrard M, Kwok IWH, Chong SZ, Teng KWW, Becht E, Chen J, Sieow JL, Penny HL, Ching GC, Devi S, Adrover JM, Li JLY, Liong KH, Tan L, Poon Z, Foo S, Chua JW, Su IH, Balabanian K, Bachelerie F, Biswas SK, Larbi A, Hwang WYK, Madan V, Koeffler HP, Wong SC, Newell EW, Hidalgo A, Ginhoux F, Ng LG. Developmental analysis of bone marrow neutrophils reveals population specialized in expansion, trafficking, and effector functions. Immunity 48: 364–379.e8, 2018. doi: 10.1016/j.immuni.2018.02.002. [DOI] [PubMed] [Google Scholar]
  • 73.Farrera C, Fadeel B. Macrophage clearance of neutrophil extracellular traps is a silent process. J Immunol 191: 2647–2656, 2013. doi: 10.4049/jimmunol.1300436. [DOI] [PubMed] [Google Scholar]
  • 74.Filep JG. Biasing the lipoxin A4/formyl peptide receptor 2 pushes inflammatory resolution. Proc Natl Acad Sci USA 110: 18033–18034, 2013. doi: 10.1073/pnas.1317798110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fine N, Hassanpour S, Borenstein A, Sima C, Oveisi M, Scholey J, Cherney D, Glogauer M. Distinct oral neutrophil subsets define health and periodontal disease states. J Dent Res 95: 931–938, 2016. doi: 10.1177/0022034516645564. [DOI] [PubMed] [Google Scholar]
  • 76.Finisguerra V, Di Conza G, Di Matteo M, Serneels J, Costa S, Thompson AA, Wauters E, Walmsley S, Prenen H, Granot Z, Casazza A, Mazzone M. MET is required for the recruitment of anti-tumoural neutrophils. Nature 522: 349–353, 2015. doi: 10.1038/nature14407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fredman G, Hellmann J, Proto JD, Kuriakose G, Colas RA, Dorweiler B, Connolly ES, Solomon R, Jones DM, Heyer EJ, Spite M, Tabas I. An imbalance between specialized pro-resolving lipid mediators and pro-inflammatory leukotrienes promotes instability of atherosclerotic plaques. Nat Commun 7: 12859, 2016. doi: 10.1038/ncomms12859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fredman G, Tabas I. Boosting inflammation resolution in atherosclerosis: The next frontiers for therapy. Am J Pathol 187: 1211–1221, 2017. doi: 10.1016/j.ajpath.2017.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Freeman SA, Goyette J, Furuya W, Woods EC, Bertozzi CR, Bergmeier W, Hinz B, van der Merwe PA, Das R, Grinstein S. Integrins form an expanding diffusion barrier that coordinates phagocytosis. Cell 164: 128–140, 2016. doi: 10.1016/j.cell.2015.11.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS, Albelda SM. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 16: 183–194, 2009. doi: 10.1016/j.ccr.2009.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fridlender ZG, Sun J, Mishalian I, Singhal S, Cheng G, Kapoor V, Horng W, Fridlender G, Bayuh R, Worthen GS, Albelda SM. Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils. PLoS One 7: e31524, 2012. doi: 10.1371/journal.pone.0031524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176: 231–241, 2007. doi: 10.1083/jcb.200606027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr, Wrobleski SK, Wakefield TW, Hartwig JH, Wagner DD. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA 107: 15880–15885, 2010. doi: 10.1073/pnas.1005743107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fuchs T, Püellmann K, Scharfenstein O, Eichner R, Stobe E, Becker A, Pechlivanidou I, Kzhyshkowska J, Gratchev A, Ganser A, Neumaier M, Beham AW, Kaminski WE. The neutrophil recombinatorial TCR-like immune receptor is expressed across the entire human life span but repertoire diversity declines in old age. Biochem Biophys Res Commun 419: 309–315, 2012. doi: 10.1016/j.bbrc.2012.02.017. [DOI] [PubMed] [Google Scholar]
  • 85.Gaipl US, Kuenkele S, Voll RE, Beyer TD, Kolowos W, Heyder P, Kalden JR, Herrmann M. Complement binding is an early feature of necrotic and a rather late event during apoptotic cell death. Cell Death Differ 8: 327–334, 2001. doi: 10.1038/sj.cdd.4400826. [DOI] [PubMed] [Google Scholar]
  • 86.Gao JL, Lee EJ, Murphy PM. Impaired antibacterial host defense in mice lacking the N-formylpeptide receptor. J Exp Med 189: 657–662, 1999. doi: 10.1084/jem.189.4.657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Garlichs CD, Eskafi S, Cicha I, Schmeisser A, Walzog B, Raaz D, Stumpf C, Yilmaz A, Bremer J, Ludwig J, Daniel WG. Delay of neutrophil apoptosis in acute coronary syndromes. J Leukoc Biol 75: 828–835, 2004. doi: 10.1189/jlb.0703358. [DOI] [PubMed] [Google Scholar]
  • 88.Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4: 891–899, 2004. doi: 10.1038/nrc1478. [DOI] [PubMed] [Google Scholar]
  • 89.Gilroy DW, Lawrence T, Perretti M, Rossi AG. Inflammatory resolution: new opportunities for drug discovery. Nat Rev Drug Discov 3: 401–416, 2004. doi: 10.1038/nrd1383. [DOI] [PubMed] [Google Scholar]
  • 90.Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol 14: 392–404, 2014. doi: 10.1038/nri3671. [DOI] [PubMed] [Google Scholar]
  • 91.Girbl T, Lenn T, Perez L, Rolas L, Barkaway A, Thiriot A, Del Fresno C, Lynam E, Hub E, Thelen M, Graham G, Alon R, Sancho D, von Andrian UH, Voisin MB, Rot A, Nourshargh S. Direct compartmentalization of the chemokines CXCL1 and CXCL2 and the atypical receptor ACKR1 determine discrete stages of neutrophil diapedesis. Immunity 49: 1062–1076.e6, 2018. doi: 10.1016/j.immuni.2018.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol 164: 1663–1667, 2000. doi: 10.4049/jimmunol.164.4.1663. [DOI] [PubMed] [Google Scholar]
  • 93.Gordon S. Phagocytosis: an immunobiologic process. Immunity 44: 463–475, 2016. doi: 10.1016/j.immuni.2016.02.026. [DOI] [PubMed] [Google Scholar]
  • 94.Greenlee-Wacker MC, Kremserová S, Nauseef WM. Lysis of human neutrophils by community-associated methicillin-resistant Staphylococcus aureus. Blood 129: 3237–3244, 2017. doi: 10.1182/blood-2017-02-766253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Greenlee-Wacker MC, Rigby KM, Kobayashi SD, Porter AR, DeLeo FR, Nauseef WM. Phagocytosis of Staphylococcus aureus by human neutrophils prevents macrophage efferocytosis and induces programmed necrosis. J Immunol 192: 4709–4717, 2014. doi: 10.4049/jimmunol.1302692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Guido BC, Zanatelli M, Tavares-de-Lima W, Oliani SM, Damazo AS. Annexin-A1 peptide down-regulates the leukocyte recruitment and up-regulates interleukin-10 release into lung after intestinal ischemia-reperfusion in mice. J Inflamm (Lond) 10: 10, 2013. doi: 10.1186/1476-9255-10-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Guo RF, Ward PA. Role of C5a in inflammatory responses. Annu Rev Immunol 23: 821–852, 2005. doi: 10.1146/annurev.immunol.23.021704.115835. [DOI] [PubMed] [Google Scholar]
  • 98.Gupta S, Kaplan MJ. The role of neutrophils and NETosis in autoimmune and renal diseases. Nat Rev Nephrol 12: 402–413, 2016. doi: 10.1038/nrneph.2016.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hacbarth E, Kajdacsy-Balla A. Low density neutrophils in patients with systemic lupus erythematosus, rheumatoid arthritis, and acute rheumatic fever. Arthritis Rheum 29: 1334–1342, 1986. doi: 10.1002/art.1780291105. [DOI] [PubMed] [Google Scholar]
  • 100.Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zychlinsky A, Waldmann H. Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol 7: 75–77, 2011. doi: 10.1038/nchembio.496. [DOI] [PubMed] [Google Scholar]
  • 101.Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci USA 107: 9813–9818, 2010. doi: 10.1073/pnas.0909927107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hampton HR, Bailey J, Tomura M, Brink R, Chtanova T. Microbe-dependent lymphatic migration of neutrophils modulates lymphocyte proliferation in lymph nodes. Nat Commun 6: 7139, 2015. doi: 10.1038/ncomms8139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hampton HR, Chtanova T. The lymph node neutrophil. Semin Immunol 28: 129–136, 2016. doi: 10.1016/j.smim.2016.03.008. [DOI] [PubMed] [Google Scholar]
  • 104.Hartl D, Krauss-Etschmann S, Koller B, Hordijk PL, Kuijpers TW, Hoffmann F, Hector A, Eber E, Marcos V, Bittmann I, Eickelberg O, Griese M, Roos D. Infiltrated neutrophils acquire novel chemokine receptor expression and chemokine responsiveness in chronic inflammatory lung diseases. J Immunol 181: 8053–8067, 2008. doi: 10.4049/jimmunol.181.11.8053. [DOI] [PubMed] [Google Scholar]
  • 105.Hartl D, Latzin P, Hordijk P, Marcos V, Rudolph C, Woischnik M, Krauss-Etschmann S, Koller B, Reinhardt D, Roscher AA, Roos D, Griese M. Cleavage of CXCR1 on neutrophils disables bacterial killing in cystic fibrosis lung disease. Nat Med 13: 1423–1430, 2007. doi: 10.1038/nm1690. [DOI] [PubMed] [Google Scholar]
  • 106.Hasturk H, Kantarci A, Ohira T, Arita M, Ebrahimi N, Chiang N, Petasis NA, Levy BD, Serhan CN, Van Dyke TE. RvE1 protects from local inflammation and osteoclast- mediated bone destruction in periodontitis. FASEB J 20: 401–403, 2006. doi: 10.1096/fj.05-4724fje. [DOI] [PubMed] [Google Scholar]
  • 107.Haworth O, Cernadas M, Yang R, Serhan CN, Levy BD. Resolvin E1 regulates interleukin 23, interferon-gamma and lipoxin A4 to promote the resolution of allergic airway inflammation. Nat Immunol 9: 873–879, 2008. doi: 10.1038/ni.1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.He R, Sang H, Ye RD. Serum amyloid A induces IL-8 secretion through a G protein-coupled receptor, FPRL1/LXA4R. Blood 101: 1572–1581, 2003. doi: 10.1182/blood-2002-05-1431. [DOI] [PubMed] [Google Scholar]
  • 109.Hellebrekers P, Hietbrink F, Vrisekoop N, Leenen LPH, Koenderman L. Neutrophil functional heterogeneity: identification of competitive phagocytosis. Front Immunol 8: 1498, 2017. doi: 10.3389/fimmu.2017.01498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Honda M, Takeichi T, Hashimoto S, Yoshii D, Isono K, Hayashida S, Ohya Y, Yamamoto H, Sugawara Y, Inomata Y. Intravital imaging of neutrophil recruitment reveals the efficacy of FPR1 blockade in hepatic ischemia-reperfusion injury. J Immunol 198: 1718–1728, 2017. doi: 10.4049/jimmunol.1601773. [DOI] [PubMed] [Google Scholar]
  • 111.Hoodless LJ, Lucas CD, Duffin R, Denvir MA, Haslett C, Tucker CS, Rossi AG. Genetic and pharmacological inhibition of CDK9 drives neutrophil apoptosis to resolve inflammation in zebrafish in vivo. Sci Rep 5: 36980, 2016. doi: 10.1038/srep36980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hu N, Mora-Jensen H, Theilgaard-Mönch K, Doornbos-van der Meer B, Huitema MG, Stegeman CA, Heeringa P, Kallenberg CG, Westra J. Differential expression of granulopoiesis related genes in neutrophil subsets distinguished by membrane expression of CD177. PLoS One 9: e99671, 2014. doi: 10.1371/journal.pone.0099671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hu N, Westra J, Huitema MG, Bijl M, Brouwer E, Stegeman CA, Heeringa P, Limburg PC, Kallenberg CG. Coexpression of CD177 and membrane proteinase 3 on neutrophils in antineutrophil cytoplasmic autoantibody-associated systemic vasculitis: anti-proteinase 3-mediated neutrophil activation is independent of the role of CD177-expressing neutrophils. Arthritis Rheum 60: 1548–1557, 2009. doi: 10.1002/art.24442. [DOI] [PubMed] [Google Scholar]
  • 114.Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell 110: 673–687, 2002. doi: 10.1016/S0092-8674(02)00971-6. [DOI] [PubMed] [Google Scholar]
  • 115.Ingersoll SA, Laval J, Forrest OA, Preininger M, Brown MR, Arafat D, Gibson G, Tangpricha V, Tirouvanziam R. Mature cystic fibrosis airway neutrophils suppress T cell function: evidence for a role of arginase 1 but not programmed death-ligand 1. J Immunol 194: 5520–5528, 2015. doi: 10.4049/jimmunol.1500312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jablonska J, Leschner S, Westphal K, Lienenklaus S, Weiss S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest 120: 1151–1164, 2010. doi: 10.1172/JCI37223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jaeger BN, Donadieu J, Cognet C, Bernat C, Ordoñez-Rueda D, Barlogis V, Mahlaoui N, Fenis A, Narni-Mancinelli E, Beaupain B, Bellanné-Chantelot C, Bajénoff M, Malissen B, Malissen M, Vivier E, Ugolini S. Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis. J Exp Med 209: 565–580, 2012. doi: 10.1084/jem.20111908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Janols H, Bergenfelz C, Allaoui R, Larsson AM, Rydén L, Björnsson S, Janciauskiene S, Wullt M, Bredberg A, Leandersson K. A high frequency of MDSCs in sepsis patients, with the granulocytic subtype dominating in gram-positive cases. J Leukoc Biol 96: 685–693, 2014. doi: 10.1189/jlb.5HI0214-074R. [DOI] [PubMed] [Google Scholar]
  • 119.Jones CN, Dimisko L, Forrest K, Judice K, Poznansky MC, Markmann JF, Vyas JM, Irimia D. Human neutrophils are primed by chemoattractant gradients for blocking the growth of Aspergillus fumigatus. J Infect Dis 213: 465–475, 2016. doi: 10.1093/infdis/jiv419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Jonsson H, Allen P, Peng SL. Inflammatory arthritis requires Foxo3a to prevent Fas ligand-induced neutrophil apoptosis. Nat Med 11: 666–671, 2005. doi: 10.1038/nm1248. [DOI] [PubMed] [Google Scholar]
  • 121.Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med 23: 279–287, 2017. doi: 10.1038/nm.4294. [DOI] [PubMed] [Google Scholar]
  • 122.József L, Khreiss T, Filep JG. CpG motifs in bacterial DNA delay apoptosis of neutrophil granulocytes. FASEB J 18: 1776–1778, 2004. doi: 10.1096/fj.04-2048fje. [DOI] [PubMed] [Google Scholar]
  • 123.József L, Zouki C, Petasis NA, Serhan CN, Filep JG. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-κ B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci USA 99: 13266–13271, 2002. doi: 10.1073/pnas.202296999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kamenyeva O, Boularan C, Kabat J, Cheung GY, Cicala C, Yeh AJ, Chan JL, Periasamy S, Otto M, Kehrl JH. Neutrophil recruitment to lymph nodes limits local humoral response to Staphylococcus aureus. PLoS Pathog 11: e1004827, 2015. doi: 10.1371/journal.ppat.1004827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kaplan A, Lee MW, Wolf AJ, Limon JJ, Becker CA, Ding M, Murali R, Lee EY, Liu GY, Wong GCL, Underhill DM. Direct antimicrobial activity of IFN-β. J Immunol 198: 4036–4045, 2017. doi: 10.4049/jimmunol.1601226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Karp CL, Flick LM, Park KW, Softic S, Greer TM, Keledjian R, Yang R, Uddin J, Guggino WB, Atabani SF, Belkaid Y, Xu Y, Whitsett JA, Accurso FJ, Wills-Karp M, Petasis NA. Defective lipoxin-mediated anti-inflammatory activity in the cystic fibrosis airway. Nat Immunol 5: 388–392, 2004. doi: 10.1038/ni1056. [DOI] [PubMed] [Google Scholar]
  • 127.Kastenmüller W, Torabi-Parizi P, Subramanian N, Lämmermann T, Germain RN. A spatially-organized multicellular innate immune response in lymph nodes limits systemic pathogen spread. Cell 150: 1235–1248, 2012. doi: 10.1016/j.cell.2012.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Keel M, Ungethüm U, Steckholzer U, Niederer E, Hartung T, Trentz O, Ertel W. Interleukin-10 counterregulates proinflammatory cytokine-induced inhibition of neutrophil apoptosis during severe sepsis. Blood 90: 3356–3363, 1997. doi: 10.1182/blood.V90.9.3356. [DOI] [PubMed] [Google Scholar]
  • 129.Kenny EF, Herzig A, Krüger R, Muth A, Mondal S, Thompson PR, Brinkmann V, Bernuth HV, Zychlinsky A. Diverse stimuli engage different neutrophil extracellular trap pathways. eLife 6: e24437, 2017. doi: 10.7554/eLife.24437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Keshari RS, Verma A, Barthwal MK, Dikshit M. Reactive oxygen species-induced activation of ERK and p38 MAPK mediates PMA-induced NETs release from human neutrophils. J Cell Biochem 114: 532–540, 2013. doi: 10.1002/jcb.24391. [DOI] [PubMed] [Google Scholar]
  • 131.Khandpur R, Carmona-Rivera C, Vivekanandan-Giri A, Gizinski A, Yalavarthi S, Knight JS, Friday S, Li S, Patel RM, Subramanian V, Thompson P, Chen P, Fox DA, Pennathur S, Kaplan MJ. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med 5: 178ra40, 2013. doi: 10.1126/scitranslmed.3005580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Khatib-Massalha E, Bhattacharya S, Massalha H, Biram A, Golan K, Kollet O, Kumari A, Avemaria F, Petrovich-Kopitman E, Gur-Cohen S, Itkin T, Brandenburger I, Spiegel A, Shulman Z, Gerhart-Hines Z, Itzkovitz S, Gunzer M, Offermanns S, Alon R, Ariel A, Lapidot T. Lactate released by inflammatory bone marrow neutrophils induces their mobilization via endothelial GPR81 signaling. Nat Commun 11: 3547, 2020. doi: 10.1038/s41467-020-17402-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kienle K, Lämmermann T. Neutrophil swarming: an essential process of the neutrophil tissue response. Immunol Rev 273: 76–93, 2016. doi: 10.1111/imr.12458. [DOI] [PubMed] [Google Scholar]
  • 134.Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol 77: 598–625, 2005. doi: 10.1189/jlb.1204697. [DOI] [PubMed] [Google Scholar]
  • 135.Koedel U, Frankenberg T, Kirschnek S, Obermaier B, Häcker H, Paul R, Häcker G. Apoptosis is essential for neutrophil functional shutdown and determines tissue damage in experimental pneumococcal meningitis. PLoS Pathog 5: e1000461, 2009. doi: 10.1371/journal.ppat.1000461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 13: 159–175, 2013. doi: 10.1038/nri3399. [DOI] [PubMed] [Google Scholar]
  • 137.Köstlin N, Kugel H, Spring B, Leiber A, Marmé A, Henes M, Rieber N, Hartl D, Poets CF, Gille C. Granulocytic myeloid derived suppressor cells expand in human pregnancy and modulate T-cell responses. Eur J Immunol 44: 2582–2591, 2014. doi: 10.1002/eji.201344200. [DOI] [PubMed] [Google Scholar]
  • 138.Kourtzelis I, Hajishengallis G, Chavakis T. Phagocytosis of apoptotic cells in resolution of inflammation. Front Immunol 11: 553, 2020. doi: 10.3389/fimmu.2020.00553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kourtzelis I, Li X, Mitroulis I, Grosser D, Kajikawa T, Wang B, Grzybek M, von Renesse J, Czogalla A, Troullinaki M, Ferreira A, Doreth C, Ruppova K, Chen LS, Hosur K, Lim JH, Chung KJ, Grossklaus S, Tausche AK, Joosten LAB, Moutsopoulos NM, Wielockx B, Castrillo A, Korostoff JM, Coskun Ü, Hajishengallis G, Chavakis T. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat Immunol 20: 40–49, 2019. doi: 10.1038/s41590-018-0249-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kreisel D, Nava RG, Li W, Zinselmeyer BH, Wang B, Lai J, Pless R, Gelman AE, Krupnick AS, Miller MJ. In vivo two-photon imaging reveals monocyte-dependent neutrophil extravasation during pulmonary inflammation. Proc Natl Acad Sci USA 107: 18073–18078, 2010. doi: 10.1073/pnas.1008737107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kremserova S, Nauseef WM. Frontline Science: Staphylococcus aureus promotes receptor-interacting protein kinase 3- and protease-dependent production of IL-1β in human neutrophils. J Leukoc Biol 105: 437–447, 2019. doi: 10.1002/JLB.4HI0918-346R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Krishnamoorthy N, Abdulnour RE, Walker KH, Engstrom BD, Levy BD. Specialized pro-resolving mediators in innate and adaptive immune responses in airway diseases. Physiol Rev 98: 1335–1370, 2018. doi: 10.1152/physrev.00026.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kruger P, Saffarzadeh M, Weber AN, Rieber N, Radsak M, von Bernuth H, Benarafa C, Roos D, Skokowa J, Hartl D. Neutrophils: Between host defence, immune modulation, and tissue injury. PLoS Pathog 11: e1004651, 2015. doi: 10.1371/journal.ppat.1004651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kuckleburg CJ, Tilkens SB, Santoso S, Newman PJ. Proteinase 3 contributes to transendothelial migration of NB1-positive neutrophils. J Immunol 188: 2419–2426, 2012. doi: 10.4049/jimmunol.1102540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Lahoz-Beneytez J, Elemans M, Zhang Y, Ahmed R, Salam A, Block M, Niederalt C, Asquith B, Macallan D. Human neutrophil kinetics: modeling of stable isotope labeling data supports short blood neutrophil half-lives. Blood 127: 3431–3438, 2016. doi: 10.1182/blood-2016-03-700336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Lambris JD, Ricklin D, Geisbrecht BV. Complement evasion by human pathogens. Nat Rev Microbiol 6: 132–142, 2008. doi: 10.1038/nrmicro1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lämmermann T. In the eye of the neutrophil swarm-navigation signals that bring neutrophils together in inflamed and infected tissues. J Leukoc Biol 100: 55–63, 2016. doi: 10.1189/jlb.1MR0915-403. [DOI] [PubMed] [Google Scholar]
  • 148.Lämmermann T, Afonso PV, Angermann BR, Wang JM, Kastenmüller W, Parent CA, Germain RN. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 498: 371–375, 2013. doi: 10.1038/nature12175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Lau D, Mollnau H, Eiserich JP, Freeman BA, Daiber A, Gehling UM, Brümmer J, Rudolph V, Münzel T, Heitzer T, Meinertz T, Baldus S. Myeloperoxidase mediates neutrophil activation by association with CD11b/CD18 integrins. Proc Natl Acad Sci USA 102: 431–436, 2005. doi: 10.1073/pnas.0405193102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Law HL, Wright RD, Iqbal AJ, Norling LV, Cooper D. A pro-resolving role for galectin-1 in acute inflammation. Front Pharmacol 11: 274, 2020. doi: 10.3389/fphar.2020.00274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lazzaretto B, Fadeel B. Intra- and extracellular degradation of neutrophil extracellular traps by macrophages and dendritic cells. J Immunol 203: 2276–2290, 2019. doi: 10.4049/jimmunol.1800159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Lee A, Whyte MKB, Haslett C. Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators. J Leukoc Biol 54: 283–288, 1993. doi: 10.1002/jlb.54.4.283. [DOI] [PubMed] [Google Scholar]
  • 153.Lefrançais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight 3: e98178, 2018. doi: 10.1172/jci.insight.98178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Leliefeld PHC, Wessels CM, Leenen LPH, Koenderman L, Pillay J. The role of neutrophils in immune dysfunction during severe inflammation. Crit Care 20: 73, 2016. doi: 10.1186/s13054-016-1250-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid mediator class switching during acute inflammation: signals in resolution. Nat Immunol 2: 612–619, 2001. doi: 10.1038/89759. [DOI] [PubMed] [Google Scholar]
  • 156.Levy BD, De Sanctis GT, Devchand PR, Kim E, Ackerman K, Schmidt BA, Szczeklik W, Drazen JM, Serhan CN. Multi-pronged inhibition of airway hyper-responsiveness and inflammation by lipoxin A4. Nat Med 8: 1018–1023, 2002. doi: 10.1038/nm748. [DOI] [PubMed] [Google Scholar]
  • 157.Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 7: 678–689, 2007. doi: 10.1038/nri2156. [DOI] [PubMed] [Google Scholar]
  • 158.Li H, Zhou X, Tan H, Hu Y, Zhang L, Liu S, Dai M, Li Y, Li Q, Mao Z, Pan P, Su X, Hu C. Neutrophil extracellular traps contribute to the pathogenesis of acid-aspiration-induced ALI/ARDS. Oncotarget 9: 1772–1784, 2018. doi: 10.18632/oncotarget.22744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev 99: 1223–1248, 2019. doi: 10.1152/physrev.00012.2018. [DOI] [PubMed] [Google Scholar]
  • 160.Lim LHK, Solito E, Russo-Marie F, Flower RJ, Perretti M. Promoting detachment of neutrophils adherent to murine postcapillary venules to control inflammation: effect of lipocortin 1. Proc Natl Acad Sci USA 95: 14535–14539, 1998. doi: 10.1073/pnas.95.24.14535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Liu M, Chen K, Yoshimura T, Liu Y, Gong W, Wang A, Gao JL, Murphy PM, Wang JM. Formylpeptide receptors are critical for rapid neutrophil mobilization in host defense against Listeria monocytogenes. Sci Rep 2: 786, 2012. doi: 10.1038/srep00786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Liu W, Yan M, Liu Y, McLeish KR, Coleman WG Jr, Rodgers GP. Olfactomedin 4 inhibits cathepsin C-mediated protease activities, thereby modulating neutrophil killing of Staphylococcus aureus and Escherichia coli in mice. J Immunol 189: 2460–2467, 2012. doi: 10.4049/jimmunol.1103179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Lokwani R, Wark PAB, Baines KJ, Barker D, Simpson JL. Hypersegmented airway neutrophils and its association with reduced lung function in adults with obstructive airway disease: an exploratory study. BMJ Open 9: e024330, 2019. doi: 10.1136/bmjopen-2018-024330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Lumbroso D, Soboh S, Maimon A, Schif-Zuck S, Ariel A, Burstyn-Cohen T. Macrophage-derived protein S facilitates apoptotic polymorphonuclear cell clearance by resolution phase macrophages and supports their reprogramming. Front Immunol 9: 358, 2018. doi: 10.3389/fimmu.2018.00358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lutaty A, Soboh S, Schif-Zuck S, Zeituni-Timor O, Rostoker R, Podolska MJ, Schauer C, Herrmann M, Muñoz LE, Ariel A. A 17-kDa fragment of lactoferrin associates with the termination of inflammation and peptides within promote resolution. Front Immunol 9: 644, 2018. doi: 10.3389/fimmu.2018.00644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Macanovic M, Sinicropi D, Shak S, Baughman S, Thiru S, Lachmann PJ. The treatment of systemic lupus erythematosus (SLE) in NZB/W F1 hybrid mice; studies with recombinant murine DNase and with dexamethasone. Clin Exp Immunol 106: 243–252, 1996. doi: 10.1046/j.1365-2249.1996.d01-839.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Maderna P, Yona S, Perretti M, Godson C. Modulation of phagocytosis of apoptotic neutrophils by supernatant from dexamethasone-treated macrophages and annexin-derived peptide Ac(2-26). J Immunol 174: 3727–3733, 2005. doi: 10.4049/jimmunol.174.6.3727. [DOI] [PubMed] [Google Scholar]
  • 168.Maimon N, Zamir ZZ, Kalkar P, Zeytuni-Timor O, Schif-Zuck S, Larisch S, Ariel A. The pro-apoptotic ARTS protein promotes neutrophil apoptosis, efferocytosis and macrophage reprogramming in resolving inflammation. Apoptosis 25: 558–573, 2020. doi: 10.1007/s10495-020-01615-3. [DOI] [PubMed] [Google Scholar]
  • 169.Malawista SE, de Boisfleury Chevance A, van Damme J, Serhan CN. Tonic inhibition of chemotaxis in human plasma. Proc Natl Acad Sci USA 105: 17949–17954, 2008. doi: 10.1073/pnas.0802572105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Malawista SE, de Boisfleury AC, Naccache PH. Inflammatory gout: observations over a half-century. FASEB J 25: 4073–4078, 2011. doi: 10.1096/fj.11-1201ufm. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Mancuso F, Flower RJ, Perretti M. Leukocyte transmigration, but not rolling or adhesion, is selectively inhibited by dexamethasone in the hamster post-capillary venule. Involvement of endogenous lipocortin 1. J Immunol 155: 377–386, 1995. [PubMed] [Google Scholar]
  • 172.Manfredi AA, Ramirez GA, Rovere-Querini P, Maugeri N. The neutrophil’s choice: phagocytose vs make neutrophil extracellular traps. Front Immunol 9: 288, 2018. doi: 10.3389/fimmu.2018.00288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Marini O, Costa S, Bevilacqua D, Calzetti F, Tamassia N, Spina C, De Sabata D, Tinazzi E, Lunardi C, Scupoli MT, Cavallini C, Zoratti E, Tinazzi I, Marchetta A, Vassanelli A, Cantini M, Gandini G, Ruzzenente A, Guglielmi A, Missale F, Vermi W, Tecchio C, Cassatella MA, Scapini P. Mature CD10+ and immature CD1 neutrophils present in G-CSF-treated donors display opposite effects on T cells. Blood 129: 1343–1356, 2017. [Erratum in Blood 129: 3271, 2017]. doi: 10.1182/blood-2016-04-713206. [DOI] [PubMed] [Google Scholar]
  • 174.Martin C, Burdon PC, Bridger G, Gutierrez-Ramos JC, Williams TJ, Rankin SM. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity 19: 583–593, 2003. doi: 10.1016/S1074-7613(03)00263-2. [DOI] [PubMed] [Google Scholar]
  • 175.Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S, Goosmann C, Brinkmann V, Lorenz M, Bidzhekov K, Khandagale AB, Konrad I, Kennerknecht E, Reges K, Holdenrieder S, Braun S, Reinhardt C, Spannagl M, Preissner KT, Engelmann B. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med 16: 887–896, 2010. doi: 10.1038/nm.2184. [DOI] [PubMed] [Google Scholar]
  • 176.Mathias JR, Perrin BJ, Liu TX, Kanki J, Look AT, Huttenlocher A. Resolution of inflammation by retrograde chemotaxis of neutrophils in transgenic zebrafish. J Leukoc Biol 80: 1281–1288, 2006. doi: 10.1189/jlb.0506346. [DOI] [PubMed] [Google Scholar]
  • 177.Matthijsen RA, Huugen D, Hoebers NT, de Vries B, Peutz-Kootstra CJ, Aratani Y, Daha MR, Tervaert JW, Buurman WA, Heeringa P. Myeloperoxidase is critically involved in the induction of organ damage after renal ischemia reperfusion. Am J Pathol 171: 1743–1752, 2007. doi: 10.2353/ajpath.2007.070184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Matute-Bello G, Liles WC, Radella F II, Steinberg KP, Ruzinski JT, Jonas M, Chi EY, Hudson LD, Martin TR. Neutrophil apoptosis in the acute respiratory distress syndrome. Am J Respir Crit Care Med 156: 1969–1977, 1997. doi: 10.1164/ajrccm.156.6.96-12081. [DOI] [PubMed] [Google Scholar]
  • 179.Maueröder C, Mahajan A, Paulus S, Gößwein S, Hahn J, Kienhöfer D, Biermann MH, Tripal P, Friedrich RP, Munoz LE, Neurath MF, Becker C, Schett GA, Herrmann M, Leppkes M. Ménage-à-Trois: the ratio of bicarbonate to CO2 and the pH regulate the capacity of neutrophils to form NETs. Front Immunol 7: 583, 2016. doi: 10.3389/fimmu.2016.00583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Maugeri N, Campana L, Gavina M, Covino C, De Metrio M, Panciroli C, Maiuri L, Maseri A, D’Angelo A, Bianchi ME, Rovere-Querini P, Manfredi AA. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J Thromb Haemost 12: 2074–2088, 2014. doi: 10.1111/jth.12710. [DOI] [PubMed] [Google Scholar]
  • 181.Maugeri N, Rovere-Querini P, Evangelista V, Covino C, Capobianco A, Bertilaccio MT, Piccoli A, Totani L, Cianflone D, Maseri A, Manfredi AA. Neutrophils phagocytose activated platelets in vivo: a phosphatidylserine, P-selectin, and β2 integrin-dependent cell clearance program. Blood 113: 5254–5265, 2009. doi: 10.1182/blood-2008-09-180794. [DOI] [PubMed] [Google Scholar]
  • 182.Mayadas TN, Cullere X. Neutrophil β2 integrins: moderators of life or death decisions. Trends Immunol 26: 388–395, 2005. doi: 10.1016/j.it.2005.05.002. [DOI] [PubMed] [Google Scholar]
  • 183.McArthur S, Gobbetti T, Kusters DH, Reutelingsperger CP, Flower RJ, Perretti M. Definition of a novel pathway centered on lysophosphatidic acid to recruit monocytes during the resolution phase of tissue inflammation. J Immunol 195: 1139–1151, 2015. doi: 10.4049/jimmunol.1500733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC, Beck PL, Muruve DA, Kubes P. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330: 362–366, 2010. doi: 10.1126/science.1195491. [DOI] [PubMed] [Google Scholar]
  • 185.Metzler KD, Goosmann C, Lubojemska A, Zychlinsky A, Papayannopoulos V. A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Reports 8: 883–896, 2014. doi: 10.1016/j.celrep.2014.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Mevorach D, Mascarenhas JO, Gershov D, Elkon KB. Complement-dependent clearance of apoptotic cells by human macrophages. J Exp Med 188: 2313–2320, 1998. doi: 10.1084/jem.188.12.2313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Mollnes TE, Brekke OL, Fung M, Fure H, Christiansen D, Bergseth G, Videm V, Lappegård KT, Köhl J, Lambris JD. Essential role of the C5a receptor in E coli-induced oxidative burst and phagocytosis revealed by a novel lepirudin-based human whole blood model of inflammation. Blood 100: 1869–1877, 2002. [PubMed] [Google Scholar]
  • 188.Molloy MJ, Grainger JR, Bouladoux N, Hand TW, Koo LY, Naik S, Quinones M, Dzutsev AK, Gao JL, Trinchieri G, Murphy PM, Belkaid Y. Intraluminal containment of commensal outgrowth in the gut during infection-induced dysbiosis. Cell Host Microbe 14: 318–328, 2013. doi: 10.1016/j.chom.2013.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Moriceau S, Lenoir G, Witko-Sarsat V. In cystic fibrosis homozygotes and heterozygotes, neutrophil apoptosis is delayed and modulated by diamide or roscovitine: evidence for an innate neutrophil disturbance. J Innate Immun 2: 260–266, 2010. doi: 10.1159/000295791. [DOI] [PubMed] [Google Scholar]
  • 190.Morioka S, Maueröder C, Ravichandran KS. Living on the edge: efferocytosis at the interface of homeostasis and pathology. Immunity 50: 1149–1162, 2019. doi: 10.1016/j.immuni.2019.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Morioka S, Perry JSA, Raymond MH, Medina CB, Zhu Y, Zhao L, Serbulea V, Onengut-Gumuscu S, Leitinger N, Kucenas S, Rathmell JC, Makowski L, Ravichandran KS. Efferocytosis induces a novel SLC program to promote glucose uptake and lactate release. Nature 563: 714–718, 2018. doi: 10.1038/s41586-018-0735-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Morita M, Kuba K, Ichikawa A, Nakayama M, Katahira J, Iwamoto R, Watanebe T, Sakabe S, Daidoji T, Nakamura S, Kadowaki A, Ohto T, Nakanishi H, Taguchi R, Nakaya T, Murakami M, Yoneda Y, Arai H, Kawaoka Y, Penninger JM, Arita M, Imai Y. The lipid mediator protectin D1 inhibits influenza virus replication and improves severe influenza. Cell 153: 112–125, 2013. doi: 10.1016/j.cell.2013.02.027. [DOI] [PubMed] [Google Scholar]
  • 193.Moses K, Brandau S. Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Semin Immunol 28: 187–196, 2016. doi: 10.1016/j.smim.2016.03.018. [DOI] [PubMed] [Google Scholar]
  • 194.Nagase H, Miyamasu M, Yamaguchi M, Imanishi M, Tsuno NH, Matsushima K, Yamamoto K, Morita Y, Hirai K. Cytokine-mediated regulation of CXCR4 expression in human neutrophils. J Leukoc Biol 71: 711–717, 2002. [PubMed] [Google Scholar]
  • 195.Naghavi M, John R, Naguib S, Siadaty MS, Grasu R, Kurian KC, van Winkle WB, Soller B, Litovsky S, Madjid M, Willerson JT, Casscells W. pH Heterogeneity of human and rabbit atherosclerotic plaques; a new insight into detection of vulnerable plaque. Atherosclerosis 164: 27–35, 2002. doi: 10.1016/S0021-9150(02)00018-7. [DOI] [PubMed] [Google Scholar]
  • 196.Nassar M, Tabib Y, Capucha T, Mizraji G, Nir T, Pevsner-Fischer M, Zilberman-Schapira G, Heyman O, Nussbaum G, Bercovier H, Wilensky A, Elinav E, Burstyn-Cohen T, Hovav AH. GAS6 is a key homeostatic immunological regulator of host-commensal interactions in the oral mucosa. Proc Natl Acad Sci USA 114: E337–E346, 2017. doi: 10.1073/pnas.1614926114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Nathan C, Ding A. Nonresolving inflammation. Cell 140: 871–882, 2010. doi: 10.1016/j.cell.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 198.Nauseef WM. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev 219: 88–102, 2007. doi: 10.1111/j.1600-065X.2007.00550.x. [DOI] [PubMed] [Google Scholar]
  • 199.Nauseef WM, Borregaard N. Neutrophils at work. Nat Immunol 15: 602–611, 2014. doi: 10.1038/ni.2921. [DOI] [PubMed] [Google Scholar]
  • 200.Nepal S, Tiruppathi C, Tsukasaki Y, Farahany J, Mittal M, Rehman J, Prockop DJ, Malik AB. STAT6 induces expression of Gas6 in macrophages to clear apoptotic neutrophils and resolve inflammation. Proc Natl Acad Sci USA 116: 16513–16518, 2019. doi: 10.1073/pnas.1821601116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Neumann A, Völlger L, Berends ET, Molhoek EM, Stapels DA, Midon M, Friães A, Pingoud A, Rooijakkers SH, Gallo RL, Mörgelin M, Nizet V, Naim HY, von Köckritz-Blickwede M. Novel role of the antimicrobial peptide LL-37 in the protection of neutrophil extracellular traps against degradation by bacterial nucleases. J Innate Immun 6: 860–868, 2014. doi: 10.1159/000363699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Ng LG, Qin JS, Roediger B, Wang Y, Jain R, Cavanagh LL, Smith AL, Jones CA, de Veer M, Grimbaldeston MA, Meeusen EN, Weninger W. Visualizing the neutrophil response to sterile tissue injury in mouse dermis reveals a three-phase cascade of events. J Invest Dermatol 131: 2058–2068, 2011. doi: 10.1038/jid.2011.179. [DOI] [PubMed] [Google Scholar]
  • 203.Noritomi DT, Soriano FG, Kellum JA, Cappi SB, Biselli PJ, Libório AB, Park M. Metabolic acidosis in patients with severe sepsis and septic shock: a longitudinal quantitative study. Crit Care Med 37: 2733–2739, 2009. doi: 10.1097/00003246-200910000-00009. [DOI] [PubMed] [Google Scholar]
  • 204.Norling LV, Perretti M. Proresolving lipid mediators enhance PMN-mediated bacterial clearance. Proc Natl Acad Sci USA 117: 9148–9150, 2020. doi: 10.1073/pnas.2004241117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immunity 41: 694–707, 2014. doi: 10.1016/j.immuni.2014.10.008. [DOI] [PubMed] [Google Scholar]
  • 206.Nourshargh S, Renshaw SA, Imhof BA. Reverse migration of neutrophils: Where, when, how and why? Trends Immunol 37: 273–286, 2016. doi: 10.1016/j.it.2016.03.006. [DOI] [PubMed] [Google Scholar]
  • 207.Oberst A, Dillon CP, Weinlich R, McCormick LL, Fitzgerald P, Pop C, Hakem R, Salvesen GS, Green DR. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 471: 363–367, 2011. doi: 10.1038/nature09852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Oberst A, Green DR. It cuts both ways: reconciling the dual roles of caspase 8 in cell death and survival. Nat Rev Mol Cell Biol 12: 757–763, 2011. doi: 10.1038/nrm3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Oldekamp S, Pscheidl S, Kress E, Soehnlein O, Jansen S, Pufe T, Wang JM, Tauber SC, Brandenburg LO. Lack of formyl peptide receptor 1 and 2 leads to more severe inflammation and higher mortality in mice with of pneumococcal meningitis. Immunology 143: 447–461, 2014. doi: 10.1111/imm.12324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol 18: 134–147, 2018. doi: 10.1038/nri.2017.105. [DOI] [PubMed] [Google Scholar]
  • 211.Park D, Han CZ, Elliott MR, Kinchen JM, Trampont PC, Das S, Collins S, Lysiak JJ, Hoehn KL, Ravichandran KS. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature 477: 220–224, 2011. doi: 10.1038/nature10340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Pashover-Schallinger E, Aswad M, Schif-Zuck S, Shapiro H, Singer P, Ariel A. The atypical chemokine receptor D6 controls macrophage efferocytosis and cytokine secretion during the resolution of inflammation. FASEB J 26: 3891–3900, 2012. doi: 10.1096/fj.11-194894. [DOI] [PubMed] [Google Scholar]
  • 213.Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature 517: 311–320, 2015. doi: 10.1038/nature14191. [DOI] [PubMed] [Google Scholar]
  • 214.Paunel-Görgülü A, Zörnig M, Lögters T, Altrichter J, Rabenhorst U, Cinatl J, Windolf J, Scholz M. Mcl-1-mediated impairment of the intrinsic apoptosis pathway in circulating neutrophils from critically ill patients can be overcome by Fas stimulation. J Immunol 183: 6198–6206, 2009. doi: 10.4049/jimmunol.0901264. [DOI] [PubMed] [Google Scholar]
  • 215.Pederzoli-Ribeil M, Maione F, Cooper D, Al-Kashi A, Dalli J, Perretti M, D’Acquisto F. Design and characterization of a cleavage-resistant Annexin A1 mutant to control inflammation in the microvasculature. Blood 116: 4288–4296, 2010. doi: 10.1182/blood-2010-02-270520. [DOI] [PubMed] [Google Scholar]
  • 216.Perez B, Henriquez C, Sarmiento J, Morales N, Folch H, Galesio JS, Uberti B, Morán G. Tamoxifen as a new therapeutic tool for neutrophilic lung inflammation. Respirology 21: 112–118, 2016. doi: 10.1111/resp.12664. [DOI] [PubMed] [Google Scholar]
  • 217.Perretti M, Chiang N, La M, Fierro IM, Marullo S, Getting SJ, Solito E, Serhan CN. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nat Med 8: 1296–1302, 2002. doi: 10.1038/nm786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Perretti M, D’Acquisto F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat Rev Immunol 9: 62–70, 2009. doi: 10.1038/nri2470. [DOI] [PubMed] [Google Scholar]
  • 219.Perretti M, Leroy X, Bland EJ, Montero-Melendez T. Resolution pharmacology: Opportunities for therapeutic innovation in inflammation. Trends Pharmacol Sci 36: 737–755, 2015. doi: 10.1016/j.tips.2015.07.007. [DOI] [PubMed] [Google Scholar]
  • 220.Perretti M, Solito E. Annexin 1 and neutrophil apoptosis. Biochem Soc Trans 32: 507–510, 2004. doi: 10.1042/bst0320507. [DOI] [PubMed] [Google Scholar]
  • 221.Peters NC, Egen JG, Secundino N, Debrabant A, Kimblin N, Kamhawi S, Lawyer P, Fay MP, Germain RN, Sacks D. In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies. Science 321: 970–974, 2008. doi: 10.1126/science.1159194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Peyssonnaux C, Datta V, Cramer T, Doedens A, Theodorakis EA, Gallo RL, Hurtado-Ziola N, Nizet V, Johnson RS. HIF-1α expression regulates the bactericidal capacity of phagocytes. J Clin Invest 115: 1806–1815, 2005. doi: 10.1172/JCI23865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Pillay J, den Braber I, Vrisekoop N, Kwast LM, de Boer RJ, Borghans JA, Tesselaar K, Koenderman L. In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood 116: 625–627, 2010. doi: 10.1182/blood-2010-01-259028. [DOI] [PubMed] [Google Scholar]
  • 224.Pillay J, Kamp VM, van Hoffen E, Visser T, Tak T, Lammers JW, Ulfman LH, Leenen LP, Pickkers P, Koenderman L. A subset of neutrophils in human systemic inflammation inhibits T cell responses through Mac-1. J Clin Invest 122: 327–336, 2012. doi: 10.1172/JCI57990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Pillay J, Ramakers BP, Kamp VM, Loi AL, Lam SW, Hietbrink F, Leenen LP, Tool AT, Pickkers P, Koenderman L. Functional heterogeneity and differential priming of circulating neutrophils in human experimental endotoxemia. J Leukoc Biol 88: 211–220, 2010. doi: 10.1189/jlb.1209793. [DOI] [PubMed] [Google Scholar]
  • 226.Pillay J, Tak T, Kamp VM, Koenderman L. Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci 70: 3813–3827, 2013. doi: 10.1007/s00018-013-1286-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Pizzagalli DU, Latino I, Pulfer A, Palomino-Segura M, Virgilio T, Farsakoglu Y, Krause R, Gonzalez SF. Characterization of the dynamic behavior of neutrophils following influenza vaccination. Front Immunol 10: 2621, 2019. doi: 10.3389/fimmu.2019.02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Poplimont H, Georgantzoglou A, Boulch M, Walker HA, Coombs C, Papaleonidopoulou F, Sarris M. Neutrophil swarming in damaged tissue is orchestrated by connexin-dependent calcium signals. Curr Biol 30: 2761–2776.e7, 2020. doi: 10.1016/j.cub.2020.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Pottecher J. Inhaled Dornase Alpha to Reduce Respiratory Failure After Severe Trauma (Online). https://clinicaltrials.gov/ct2/show/NCT03368092 [21 April 2020].
  • 230.Puellmann K, Kaminski WE, Vogel M, Nebe CT, Schroeder J, Wolf H, Beham AW. A variable immunoreceptor in a subpopulation of human neutrophils. Proc Natl Acad Sci USA 103: 14441–14446, 2006. doi: 10.1073/pnas.0603406103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Rajamäki K, Nordström T, Nurmi K, Åkerman KE, Kovanen PT, Öörni K, Eklund KK. Extracellular acidosis is a novel danger signal alerting innate immunity via the NLRP3 inflammasome. J Biol Chem 288: 13410–13419, 2013. doi: 10.1074/jbc.M112.426254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Rarok AA, Stegeman CA, Limburg PC, Kallenberg CG. Neutrophil membrane expression of proteinase 3 (PR3) is related to relapse in PR3-ANCA-associated vasculitis. J Am Soc Nephrol 13: 2232–2238, 2002. doi: 10.1097/01.ASN.0000028642.26222.00. [DOI] [PubMed] [Google Scholar]
  • 233.Ratanarat R, Cazzavillan S, Ricci Z, Rassu M, Segala C, de Cal M, Cruz D, Corradi V, Manfro S, Roessler E, Levin N, Ronco C. Usefulness of a molecular strategy for the detection of bacterial DNA in patients with severe sepsis undergoing continuous renal replacement therapy. Blood Purif 25: 106–111, 2007. doi: 10.1159/000096406. [DOI] [PubMed] [Google Scholar]
  • 234.Rathod KS, Kapil V, Velmurugan S, Khambata RS, Siddique U, Khan S, Van Eijl S, Gee LC, Bansal J, Pitrola K, Shaw C, D’Acquisto F, Colas RA, Marelli-Berg F, Dalli J, Ahluwalia A. Accelerated resolution of inflammation underlies sex differences in inflammatory responses in humans. J Clin Invest 127: 169–182, 2017. doi: 10.1172/JCI89429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Reátegui E, Jalali F, Khankhel AH, Wong E, Cho H, Lee J, Serhan CN, Dalli J, Elliott H, Irimia D. Microscale arrays for the profiling of start and stop signals coordinating human-neutrophil swarming. Nat Biomed Eng 1: 0094, 2017. doi: 10.1038/s41551-017-0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Reglero-Real N, Rolas L, Nourshargh S. Leukocyte trafficking: time to take time seriously. Immunity 50: 273–275, 2019. doi: 10.1016/j.immuni.2019.01.013. [DOI] [PubMed] [Google Scholar]
  • 237.Rigby DA, Ferguson DJ, Johnson LA, Jackson DG. Neutrophils rapidly transit inflamed lymphatic vessel endothelium via integrin-dependent proteolysis and lipoxin-induced junctional retraction. J Leukoc Biol 98: 897–912, 2015. doi: 10.1189/jlb.1HI0415-149R. [DOI] [PubMed] [Google Scholar]
  • 238.Rossi AG, Sawatzky DA, Walker A, Ward C, Sheldrake TA, Riley NA, Caldicott A, Martinez-Losa M, Walker TR, Duffin R, Gray M, Crescenzi E, Martin MC, Brady HJ, Savill JS, Dransfield I, Haslett C. Cyclin-dependent kinase inhibitors enhance the resolution of inflammation by promoting inflammatory cell apoptosis. Nat Med 12: 1056–1064, 2006. [Erratum in Nat Med 12: 1434, 2006]. doi: 10.1038/nm1468. [DOI] [PubMed] [Google Scholar]
  • 239.Rother N, Pieterse E, Lubbers J, Hilbrands L, van der Vlag J. Acetylated histones in apoptotic microparticles drive the formation of neutrophil extracellular traps in active lupus nephritis. Front Immunol 8: 1136, 2017. doi: 10.3389/fimmu.2017.01136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Sagiv JY, Michaeli J, Assi S, Mishalian I, Kisos H, Levy L, Damti P, Lumbroso D, Polyansky L, Sionov RV, Ariel A, Hovav AH, Henke E, Fridlender ZG, Granot Z. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Reports 10: 562–573, 2015. doi: 10.1016/j.celrep.2014.12.039. [DOI] [PubMed] [Google Scholar]
  • 241.Saha S, Biswas SK. Tumor-associated neutrophils show phenotypic and functional divergence in human lung cancer. Cancer Cell 30: 11–13, 2016. doi: 10.1016/j.ccell.2016.06.016. [DOI] [PubMed] [Google Scholar]
  • 242.Kumaran Satyanarayanan S, El Kebir D, Soboh S, Butenko S, Sekheri M, Saadi J, Peled N, Assi S, Othman A, Schif-Zuck S, Feuermann Y, Barkan D, Sher N, Filep JG, Ariel A. IFN-β is a macrophage-derived effector cytokine facilitating the resolution of bacterial inflammation. Nat Commun 10: 3471, 2019. doi: 10.1038/s41467-019-10903-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Savill J, Dransfield I, Gregory C, Haslett C. A blast from the past: clearance of apoptotic cells regulates immune responses. Nat Rev Immunol 2: 965–975, 2002. doi: 10.1038/nri957. [DOI] [PubMed] [Google Scholar]
  • 244.Savill JS, Wyllie AH, Henson JE, Walport MJ, Henson PM, Haslett C. Macrophage phagocytosis of aging neutrophils in inflammation. Programmed cell death in the neutrophil leads to its recognition by macrophages. J Clin Invest 83: 865–875, 1989. doi: 10.1172/JCI113970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Schauer C, Janko C, Munoz LE, Zhao Y, Kienhöfer D, Frey B, Lell M, Manger B, Rech J, Naschberger E, Holmdahl R, Krenn V, Harrer T, Jeremic I, Bilyy R, Schett G, Hoffmann M, Herrmann M. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med 20: 511–517, 2014. doi: 10.1038/nm.3547. [DOI] [PubMed] [Google Scholar]
  • 246.Scheiermann C, Colom B, Meda P, Patel NS, Voisin MB, Marrelli A, Woodfin A, Pitzalis C, Thiemermann C, Aurrand-Lions M, Imhof BA, Nourshargh S. Junctional adhesion molecule-C mediates leukocyte infiltration in response to ischemia reperfusion injury. Arterioscler Thromb Vasc Biol 29: 1509–1515, 2009. doi: 10.1161/ATVBAHA.109.187559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Schif-Zuck S, Gross N, Assi S, Rostoker R, Serhan CN, Ariel A. Saturated-efferocytosis generates pro-resolving CD11blow macrophages: modulation by resolvins and glucocorticoids. Eur J Immunol 41: 366–379, 2011. doi: 10.1002/eji.201040801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 447: 869–874, 2007. doi: 10.1038/nature05877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Schwartz DA, Quinn TJ, Thorne PS, Sayeed S, Yi AK, Krieg AM. CpG motifs in bacterial DNA cause inflammation in the lower respiratory tract. J Clin Invest 100: 68–73, 1997. doi: 10.1172/JCI119523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Sekheri M, El Kebir D, Edner N, Filep JG. 15-Epi-LXA4 and 17-epi-RvD1 restore TLR9-mediated impaired neutrophil phagocytosis and accelerate resolution of lung inflammation. Proc Natl Acad Sci USA 117: 7971–7980, 2020. doi: 10.1073/pnas.1920193117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Seki H, Fukunaga K, Arita M, Arai H, Nakanishi H, Taguchi R, Miyasho T, Takamiya R, Asano K, Ishizaka A, Takeda J, Levy BD. The anti-inflammatory and proresolving mediator resolvin E1 protects mice from bacterial pneumonia and acute lung injury. J Immunol 184: 836–843, 2010. doi: 10.4049/jimmunol.0901809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Semerad CL, Liu F, Gregory AD, Stumpf K, Link DC. G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood. Immunity 17: 413–423, 2002. doi: 10.1016/S1074-7613(02)00424-7. [DOI] [PubMed] [Google Scholar]
  • 253.Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510: 92–101, 2014. doi: 10.1038/nature13479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. J Exp Med 192: 1197–1204, 2000. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Serhan CN, Gotlinger K, Hong S, Lu Y, Siegelman J, Baer T, Yang R, Colgan SP, Petasis NA. Anti-inflammatory actions of neuroprotectin D1/protectin D1 and its natural stereoisomers: assignments of dihydroxy-containing docosatrienes. J Immunol 176: 1848–1859, 2006. doi: 10.4049/jimmunol.176.3.1848. [DOI] [PubMed] [Google Scholar]
  • 256.Serhan CN, Jain A, Marleau S, Clish C, Kantarci A, Behbehani B, Colgan SP, Stahl GL, Merched A, Petasis NA, Chan L, Van Dyke TE. Reduced inflammation and tissue damage in transgenic rabbits overexpressing 15-lipoxygenase and endogenous anti-inflammatory lipid mediators. J Immunol 171: 6856–6865, 2003. doi: 10.4049/jimmunol.171.12.6856. [DOI] [PubMed] [Google Scholar]
  • 257.Serhan CN, Levy BD. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J Clin Invest 128: 2657–2669, 2018. doi: 10.1172/JCI97943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol 6: 1191–1197, 2005. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 259.Serhan CN, Yang R, Martinod K, Kasuga K, Pillai PS, Porter TF, Oh SF, Spite M. Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions. J Exp Med 206: 15–23, 2009. doi: 10.1084/jem.20081880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Silvestre-Roig C, Hidalgo A, Soehnlein O. Neutrophil heterogeneity: implications for homeostasis and pathogenesis. Blood 127: 2173–2181, 2016. doi: 10.1182/blood-2016-01-688887. [DOI] [PubMed] [Google Scholar]
  • 261.Singhal S, Bhojnagarwala PS, O’Brien S, Moon EK, Garfall AL, Rao AS, Quatromoni JG, Stephen TL, Litzky L, Deshpande C, Feldman MD, Hancock WW, Conejo-Garcia JR, Albelda SM, Eruslanov EB. Origin and role of a subset of tumor-associated neutrophils with antigen-presenting cell features in early stage human lung cancer. Cancer Cell 30: 120–135, 2016. doi: 10.1016/j.ccell.2016.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Solito E, Mulla A, Morris JF, Christian HC, Flower RJ, Buckingham JC. Dexamethasone induces rapid serine-phosphorylation and membrane translocation of annexin 1 in a human folliculostellate cell line via a novel nongenomic mechanism involving the glucocorticoid receptor, protein kinase C, phosphatidylinositol 3-kinase, and mitogen-activated protein kinase. Endocrinology 144: 1164–1174, 2003. doi: 10.1210/en.2002-220592. [DOI] [PubMed] [Google Scholar]
  • 263.Sørensen OE, Borregaard N. Neutrophil extracellular traps - the dark side of neutrophils. J Clin Invest 126: 1612–1620, 2016. doi: 10.1172/JCI84538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Spite M, Norling LV, Summers L, Yang R, Cooper D, Petasis NA, Flower RJ, Perretti M, Serhan CN. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature 461: 1287–1291, 2009. doi: 10.1038/nature08541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Ssemaganda A, Kindinger L, Bergin P, Nielsen L, Mpendo J, Ssetaala A, Kiwanuka N, Munder M, Teoh TG, Kropf P, Müller I. Characterization of neutrophil subsets in healthy human pregnancies. PLoS One 9: e85696, 2014. doi: 10.1371/journal.pone.0085696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Stuqui B, de Paula-Silva M, Carlos CP, Ullah A, Arni RK, Gil CD, Oliani SM. Ac2–26 mimetic peptide of annexin A1 inhibits local and systemic inflammatory processes induced by Bothrops moojeni venom and the Lys-49 phospholipase A2 in a rat model. PLoS One 10: e0130803, 2015. doi: 10.1371/journal.pone.0130803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Sugimoto MA, Vago JP, Perretti M, Teixeira MM. Mediators of the resolution of the inflammatory response. Trends Immunol 40: 212–227, 2019. doi: 10.1016/j.it.2019.01.007. [DOI] [PubMed] [Google Scholar]
  • 268.Sun YP, Oh SF, Uddin J, Yang R, Gotlinger K, Campbell E, Colgan SP, Petasis NA, Serhan CN. Resolvin D1 and its aspirin-triggered 17R epimer. Stereochemical assignments, anti-inflammatory properties, and enzymatic inactivation. J Biol Chem 282: 9323–9334, 2007. doi: 10.1074/jbc.M609212200. [DOI] [PubMed] [Google Scholar]
  • 269.Tadie JM, Bae HB, Jiang S, Park DW, Bell CP, Yang H, Pittet JF, Tracey K, Thannickal VJ, Abraham E, Zmijewski JW. HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Physiol Lung Cell Mol Physiol 304: L342–L349, 2013. doi: 10.1152/ajplung.00151.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Tak T, Tesselaar K, Pillay J, Borghans JA, Koenderman L. What’s your age again? Determination of human neutrophil half-lives revisited. J Leukoc Biol 94: 595–601, 2013. doi: 10.1189/jlb.1112571. [DOI] [PubMed] [Google Scholar]
  • 271.Takashima A, Yao Y. Neutrophil plasticity: acquisition of phenotype and functionality of antigen-presenting cell. J Leukoc Biol 98: 489–496, 2015. doi: 10.1189/jlb.1MR1014-502R. [DOI] [PubMed] [Google Scholar]
  • 272.Tan BH, Meinken C, Bastian M, Bruns H, Legaspi A, Ochoa MT, Krutzik SR, Bloom BR, Ganz T, Modlin RL, Stenger S. Macrophages acquire neutrophil granules for antimicrobial activity against intracellular pathogens. J Immunol 177: 1864–1871, 2006. doi: 10.4049/jimmunol.177.3.1864. [DOI] [PubMed] [Google Scholar]
  • 273.Taylor PR, Roy S, Leal SM Jr, Sun Y, Howell SJ, Cobb BA, Li X, Pearlman E. Autocrine IL-17A-IL-17RC neutrophil activation in fungal infections is regulated by IL-6, IL-23, RORγt and dectin-2. Nat Immunol 15: 143–151, 2014. doi: 10.1038/ni.2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Teixeira RAP, Mimura KK, Araujo LP, Greco KV, Oliani SM. The essential role of annexin A1 mimetic peptide in the skin allograft survival. J Tissue Eng Regen Med 10: E44–E53, 2016. doi: 10.1002/term.1773. [DOI] [PubMed] [Google Scholar]
  • 275.Thieblemont N, Witko-Sarsat V, Ariel A. Regulation of macrophage activation by proteins expressed on apoptotic neutrophils: subversion towards autoimmunity by proteinase 3. Eur J Clin Invest 48, Suppl 2: e12990, 2018. doi: 10.1111/eci.12990. [DOI] [PubMed] [Google Scholar]
  • 276.Thompson AA, Elks PM, Marriott HM, Eamsamarng S, Higgins KR, Lewis A, Williams L, Parmar S, Shaw G, McGrath EE, Formenti F, Van Eeden FJ, Kinnula VL, Pugh CW, Sabroe I, Dockrell DH, Chilvers ER, Robbins PA, Percy MJ, Simon MC, Johnson RS, Renshaw SA, Whyte MK, Walmsley SR. Hypoxia-inducible factor 2α regulates key neutrophil functions in humans, mice, and zebrafish. Blood 123: 366–376, 2014. doi: 10.1182/blood-2013-05-500207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Tirouvanziam R, Gernez Y, Conrad CK, Moss RB, Schrijver I, Dunn CE, Davies ZA, Herzenberg LA, Herzenberg LA. Profound functional and signaling changes in viable inflammatory neutrophils homing to cystic fibrosis airways. Proc Natl Acad Sci USA 105: 4335–4339, 2008. doi: 10.1073/pnas.0712386105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Tourneur L, Witko-Sarsat V. Inflammasome activation: Neutrophils go their own way. J Leukoc Biol 105: 433–436, 2019. doi: 10.1002/JLB.3CE1118-433R. [DOI] [PubMed] [Google Scholar]
  • 279.Tsurumaki H, Mogi C, Aoki-Saito H, Tobo M, Kamide Y, Yatomi M, Sato K, Dobashi K, Ishizuka T, Hisada T, Yamada M, Okajima F. Protective role of proton-sensing TDAG8 in lipopolysaccharide-induced acute lung injury. Int J Mol Sci 16: 28931–28942, 2015. doi: 10.3390/ijms161226145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Uddin M, Nong G, Ward J, Seumois G, Prince LR, Wilson SJ, Cornelius V, Dent G, Djukanovic R. Prosurvival activity for airway neutrophils in severe asthma. Thorax 65: 684–689, 2010. doi: 10.1136/thx.2009.120741. [DOI] [PubMed] [Google Scholar]
  • 281.Uderhardt S, Martins AJ, Tsang JS, Lämmermann T, Germain RN. Resident macrophages cloak tissue microlesions to prevent neutrophil-driven inflammatory damage. Cell 177: 541–555.e17, 2019. doi: 10.1016/j.cell.2019.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Uhl B, Vadlau Y, Zuchtriegel G, Nekolla K, Sharaf K, Gaertner F, Massberg S, Krombach F, Reichel CA. Aged neutrophils contribute to the first line of defense in the acute inflammatory response. Blood 128: 2327–2337, 2016. doi: 10.1182/blood-2016-05-718999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Vago JP, Nogueira CR, Tavares LP, Soriani FM, Lopes F, Russo RC, Pinho V, Teixeira MM, Sousa LP. Annexin A1 modulates natural and glucocorticoid-induced resolution of inflammation by enhancing neutrophil apoptosis. J Leukoc Biol 92: 249–258, 2012. doi: 10.1189/jlb.0112008. [DOI] [PubMed] [Google Scholar]
  • 284.Vago JP, Tavares LP, Sugimoto MA, Lima GL, Galvão I, de Caux TR, Lima KM, Ribeiro AL, Carneiro FS, Nunes FF, Pinho V, Perretti M, Teixeira MM, Sousa LP. Proresolving actions of synthetic and natural protease inhibitors are mediated by annexin A1. J Immunol 196: 1922–1932, 2016. doi: 10.4049/jimmunol.1500886. [DOI] [PubMed] [Google Scholar]
  • 285.Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, Denny MF, Plumas J, Chaperot L, Kretzler M, Bruce AT, Kaplan MJ. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187: 538–552, 2011. doi: 10.4049/jimmunol.1100450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Vogel S, Bodenstein R, Chen Q, Feil S, Feil R, Rheinlaender J, Schäffer TE, Bohn E, Frick JS, Borst O, Münzer P, Walker B, Markel J, Csanyi G, Pagano PJ, Loughran P, Jessup ME, Watkins SC, Bullock GC, Sperry JL, Zuckerbraun BS, Billiar TR, Lotze MT, Gawaz M, Neal MD. Platelet-derived HMGB1 is a critical mediator of thrombosis. J Clin Invest 125: 4638–4654, 2015. doi: 10.1172/JCI81660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Wan M, Godson C, Guiry PJ, Agerberth B, Haeggström JZ. Leukotriene B4/antimicrobial peptide LL-37 proinflammatory circuits are mediated by BLT1 and FPR2/ALX and are counterregulated by lipoxin A4 and resolvin E1. FASEB J 25: 1697–1705, 2011. doi: 10.1096/fj.10-175687. [DOI] [PubMed] [Google Scholar]
  • 288.Wang SB, Hu KM, Seamon KJ, Mani V, Chen Y, Gronert K. Estrogen negatively regulates epithelial wound healing and protective lipid mediator circuits in the cornea. FASEB J 26: 1506–1516, 2012. doi: 10.1096/fj.11-198036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Wang X, He Z, Liu H, Yousefi S, Simon HU. Neutrophil necroptosis is triggered by ligation of adhesion molecules following GM-CSF priming. J Immunol 197: 4090–4100, 2016. doi: 10.4049/jimmunol.1600051. [DOI] [PubMed] [Google Scholar]
  • 290.Wang X, Yousefi S, Simon HU. Necroptosis and neutrophil-associated disorders. Cell Death Dis 9: 111, 2018. doi: 10.1038/s41419-017-0058-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Wang Y, Subramanian M, Yurdagul A Jr, Barbosa-Lorenzi VC, Cai B, de Juan-Sanz J, Ryan TA, Nomura M, Maxfield FR, Tabas I. Mitochondrial fission promotes the continued clearance of apoptotic cells by macrophages. Cell 171: 331–345.e22, 2017. doi: 10.1016/j.cell.2017.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Wang Z, Chen F, Zhai R, Zhang L, Su L, Lin X, Thompson T, Christiani DC. Plasma neutrophil elastase and elafin imbalance is associated with acute respiratory distress syndrome (ARDS) development. PLoS One 4: e4380, 2009. doi: 10.1371/journal.pone.0004380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol 18: 127–136, 2017. doi: 10.1038/nrm.2016.149. [DOI] [PubMed] [Google Scholar]
  • 294.Welin A, Amirbeagi F, Christenson K, Björkman L, Björnsdottir H, Forsman H, Dahlgren C, Karlsson A, Bylund J. The human neutrophil subsets defined by the presence or absence of OLFM4 both transmigrate into tissue in vivo and give rise to distinct NETs in vitro. PLoS One 8: e69575, 2013. doi: 10.1371/journal.pone.0069575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol 17: 363–375, 2017. doi: 10.1038/nri.2017.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Westman J, Grinstein S, Marques PE. Phagocytosis of necrotic debris at sites of injury and inflammation. Front Immunol 10: 3030, 2020. doi: 10.3389/fimmu.2019.03030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Wicki S, Gurzeler U, Wei-Lynn Wong W, Jost PJ, Bachmann D, Kaufmann T. Loss of XIAP facilitates switch to TNFα-induced necroptosis in mouse neutrophils. Cell Death Dis 7: e2422, 2016. doi: 10.1038/cddis.2016.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Wong SL, Demers M, Martinod K, Gallant M, Wang Y, Goldfine AB, Kahn CR, Wagner DD. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med 21: 815–819, 2015. doi: 10.1038/nm.3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Woodfin A, Voisin MB, Beyrau M, Colom B, Caille D, Diapouli FM, Nash GB, Chavakis T, Albelda SM, Rainger GE, Meda P, Imhof BA, Nourshargh S. The junctional adhesion molecule JAM-C regulates polarized transendothelial migration of neutrophils in vivo. Nat Immunol 12: 761–769, 2011. doi: 10.1038/ni.2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Xie Q, Klesney-Tait J, Keck K, Parlet C, Borcherding N, Kolb R, Li W, Tygrett L, Waldschmidt T, Olivier A, Chen S, Liu GH, Li X, Zhang W. Characterization of a novel mouse model with genetic deletion of CD177. Protein Cell 6: 117–126, 2015. doi: 10.1007/s13238-014-0109-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Yaseen H, Butenko S, Polishuk-Zotkin I, Schif-Zuck S, Pérez-Sáez JM, Rabinovich GA, Ariel A. Galectin-1 facilitates macrophage reprogramming and resolution of inflammation through IFN-β. Front Pharmacol 11: 901, 2020. doi: 10.3389/fphar.2020.00901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Yazid S, Leoni G, Getting SJ, Cooper D, Solito E, Perretti M, Flower RJ. Antiallergic cromones inhibit neutrophil recruitment onto vascular endothelium via annexin-A1 mobilization. Arterioscler Thromb Vasc Biol 30: 1718–1724, 2010. doi: 10.1161/ATVBAHA.110.209536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev 61: 119–161, 2009. doi: 10.1124/pr.109.001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Yipp BG, Petri B, Salina D, Jenne CN, Scott BN, Zbytnuik LD, Pittman K, Asaduzzaman M, Wu K, Meijndert HC, Malawista SE, de Boisfleury Chevance A, Zhang K, Conly J, Kubes P. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med 18: 1386–1393, 2012. doi: 10.1038/nm.2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Yousefi S, Mihalache C, Kozlowski E, Schmid I, Simon HU. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ 16: 1438–1444, 2009. doi: 10.1038/cdd.2009.96. [DOI] [PubMed] [Google Scholar]
  • 306.Yousefi S, Stojkov D, Germic N, Simon D, Wang X, Benarafa C, Simon HU. Untangling “NETosis” from NETs. Eur J Immunol 49: 221–227, 2019. doi: 10.1002/eji.201747053. [DOI] [PubMed] [Google Scholar]
  • 307.Yuen J, Pluthero FG, Douda DN, Riedl M, Cherry A, Ulanova M, Kahr WH, Palaniyar N, Licht C. NETosing neutrophils activate complement both on their own NETs and bacteria via alternative and non-alternative pathways. Front Immunol 7: 137, 2016. doi: 10.3389/fimmu.2016.00137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ, Burk RD, Kunisaki Y, Jang JE, Scheiermann C, Merad M, Frenette PS. Neutrophil ageing is regulated by the microbiome. Nature 525: 528–532, 2015. doi: 10.1038/nature15367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464: 104–107, 2010. doi: 10.1038/nature08780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Zheng W, Warner R, Ruggeri R, Su C, Cortes C, Skoura A, Ward J, Ahn K, Kalgutkar A, Sun D, Maurer TS, Bonin PD, Okerberg C, Bobrowski W, Kawabe T, Zhang Y, Coskran T, Bell S, Kapoor B, Johnson K, Buckbinder L. PF-1355, a mechanism-based myeloperoxidase inhibitor, prevents immune complex vasculitis and anti-glomerular basement membrane glomerulonephritis. J Pharmacol Exp Ther 353: 288–298, 2015. doi: 10.1124/jpet.114.221788. [DOI] [PubMed] [Google Scholar]
  • 311.Zhou L, Chong MMW, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity 30: 646–655, 2009. doi: 10.1016/j.immuni.2009.05.001. [DOI] [PubMed] [Google Scholar]
  • 312.Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, Blair C, Weber A, Barnes BJ, Egeblad M, Woods RJ, Kanthi Y, Knight JS. Neutrophil extracellular traps in COVID-19. JCI Insight 5: e138999, 2020. doi: 10.1172/jci.insight.138999. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

RESOURCES