Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2013 Dec 6;306(3):L217–L230. doi: 10.1152/ajplung.00311.2013

The mercurial nature of neutrophils: still an enigma in ARDS?

Andrew E Williams 1, Rachel C Chambers 1,
PMCID: PMC3920201  PMID: 24318116

Abstract

The acute respiratory distress syndrome (ARDS) is a life-threatening lung condition resulting from direct and indirect insults to the lung. It is characterized by disruption of the endothelial-epithelial barrier, alveolar damage, pulmonary edema, and respiratory failure. A key feature of ARDS is the accumulation of neutrophils in the lung microvasculature, interstitium, and alveolar space. Despite a clear association between neutrophil influx into the lung and disease severity, there is some debate as to whether neutrophils directly contribute to disease pathogenesis. The primary function of neutrophils is to provide immediate host defense against pathogenic microorganisms. Neutrophils release numerous antimicrobial factors such as reactive oxygen species, proteinases, and neutrophil extracellular traps. However, these factors are also toxic to host cells and can result in bystander tissue damage. The excessive accumulation of neutrophils in ARDS may therefore contribute to disease progression. Central to neutrophil recruitment is the release of chemokines, including the archetypal neutrophil chemoattractant IL-8, from resident pulmonary cells. However, the chemokine network in the inflamed lung is complex and may involve several other chemokines, including CXCL10, CCL2, and CCL7. This review will therefore focus on the experimental and clinical evidence supporting neutrophils as key players in ARDS and the chemokines involved in recruiting them into the lung.

Keywords: ARDS, inflammation, neutrophil, chemokine, lung injury


acute lung injury (ALI) and its more severe form acute respiratory distress syndrome (ARDS) are common life-threatening conditions leading to respiratory failure. The current definition of ARDS now includes ALI as mild ARDS and is based on clinical criteria regarding diagnostic timing, bilateral opacities following chest imaging, pulmonary edema, hypoxemia, and a minimal requirement for positive end-expiratory pressure applied by mechanical ventilation to maintain oxygenation (47). The severity of ARDS is classified as mild, moderate, or severe based on partial pressure of arterial oxygen to fraction of inspired oxygen ratios (PaO2/FiO2), whereby mild has <200 PaO2/FiO2 ≤ 300, moderate <100 PaO2/FiO2 ≤ 200, and severe PaO2/FiO2 ≤ 100 (47). ARDS arises from a variety of local and systemic insults, of which sepsis, pneumonia, and trauma are the most common. The incidence of ARDS is 79/100,000 per year (151) with a mortality rate of 30–65% (130). ARDS is characterized by increased alveolar-capillary barrier permeability, lung edema, impaired oxygenation, and hypoxemia. Disease pathogenesis is also associated with an increased release of several inflammatory mediators, including TNF, which can directly trigger epithelial cell death (7). Furthermore, the accumulation of neutrophils in the lung interstitium and alveolar space is a key characteristic of ARDS. The rapid development of interstitial and intra-alveolar fibrosis in ARDS can further contribute to prolonged respiratory failure and increased mortality (113). Despite some improvements with use of lung-protective ventilator strategies, both morbidity and mortality remain high.

Neutrophils are the first leukocytes to be recruited to sites of inflammation in response to chemotactic factors released by activated macrophages and pulmonary epithelial and endothelial cells (104, 180, 181). At sites of inflammation neutrophils release several antimicrobial factors such as reactive oxygen species (ROS), antimicrobial peptides, and multiple proteinases, the latter of which also help degrade the extracellular matrix during migration (57, 138), together with the formation of neutrophil extracellular traps (NETs). Although neutrophils provide a first line of defense against microbes, excessive recruitment and activation can lead to bystander tissue damage and further loss of lung function (67). Therefore neutrophils and their associated chemotactic factors are thought to significantly contribute to the progression of ARDS. This review will focus of the role that neutrophils play during ARDS and the chemokines thought to be critical for neutrophil recruitment into inflamed lung.

Contribution of Neutrophils to ARDS

The evidence that neutrophils have a direct influence on the development of ARDS comes from both human clinical data and animal studies. It has been reported that the concentration of neutrophils in the bronchoalveolar lavage fluid (BALF) of patients with ARDS correlates with the severity of disease and with poor outcome (1, 3, 115). In addition to having elevated neutrophil numbers, BALF obtained from patients with ARDS is highly chemotactic for human neutrophils compared with BALF from normal subjects (141). Leukocyte counts from the blood were reduced in patients that developed ARDS compared with at risk patients (169), further indicating that neutrophil sequestration into the lungs from the circulation is indicative of ARDS (77). The kinetics of neutrophil recruitment may also be important in disease pathogenesis, as neutrophil counts remain higher in those patients who eventually die as a result of sepsis-induced ARDS compared with survivors (162). Neutrophils isolated from sepsis patients with a confirmed clinical diagnosis of ARDS were also shown to compromise the integrity of endothelial cell monolayers in vitro (51). However, despite an obvious association between ARDS and neutrophil recruitment, it has been difficult to establish a direct causal relationship between neutrophilia and endothelial-epithelial barrier disruption or clinical outcomes in ARDS. For example, neutropenic patients can still develop ARDS, as measured by arterial hypoxemia and diffuse bilateral pulmonary infiltrates, in the absence of invading neutrophils (137). This may reflect the heterogeneity of ARDS, since both neutrophil-dependent and neutrophil-independent processes are thought to influence the progression of disease.

Although clinical data does suggest a central role for neutrophils in ARDS, the majority of studies have been performed in animal models. Neutrophil-dependent models of ARDS include LPS-induced lung injury (2, 30), acid-induced lung injury (49), ventilator-induced lung injury (89), and transfusion-related lung injury (111). Neutrophil depletion in these models ameliorates certain features of ARDS, including endothelial-epithelial cell damage and capillary-alveolar permeability, thereby supporting the notion that neutrophils are central mediators of disease pathogenesis. In a rat model of LPS-induced lung injury, treatment with steroids also reduced neutrophil infiltration and cytokine levels, resulting in a consequential attenuation of lung damage, although these findings have been poorly translated into ARDS patients (48, 136, 178). The association between neutrophil influx and barrier permeability is likely to be complex. For example, in the mouse model of LPS-induced lung injury only the initial phase of neutrophil recruitment (3 h after LPS administration) was associated with a reduction in BALF protein levels, as an indication of microvasculature leak, whereas depleting neutrophils at 24 h had no effect on BALF protein (30). This could be explained by the recruitment of different neutrophil subpopulations into the lung, since marginated neutrophils within the pulmonary microvasculature are known to have a more proinflammatory phenotype compared with newly released neutrophils from the bone marrow (50), the former accounting for the initial infiltrate and the latter the secondary infiltrate.

In contrast, oleic acid-induced and hyperoxia-induced models of ARDS are independent of neutrophil recruitment (67), since depletion of neutrophils does not prevent capillary-alveolar leak (75, 143). Because of the conflicting findings between various animal models it is still unclear whether the actual process of neutrophil recruitment into the lungs is sufficient to cause endothelial-epithelial barrier disruption on its own (41). Indeed, the recruitment of neutrophils into normal human lung, in response to the leukotriene LTB4, did not cause significant alveolar epithelial permeability (114). It therefore seems likely that neutrophil activation is required to cause bystander tissue damage (156) and that several factors combine to induce lung injury (Fig. 1), through the release of proinflammatory cytokines, the generation of ROS or free radicals, mechanical stress to the endothelium or epithelium, and the release of neutrophil proteinases or other host defense proteins (13, 85). Similar events are also likely to occur in human ARDS, to varying degrees, depending on individual responses, the cause of initial injury, and the involvement of various microorganisms, including Streptococcus pneumoniae (116).

Fig. 1.

Fig. 1.

Role of neutrophils in acute respiratory distress syndrome (ARDS). An initial inflammatory insult to the lung results in the increased expression and release of proinflammatory cytokines, such as IL-1β, TNF, and IL-6, and chemokines, such as IL-8 (CXCL8) and CCL2 (MCP-1). This results in the activation and recruitment of neutrophils into areas of inflamed lung. Activated neutrophils are capable of releasing chemokines that enhance leukocyte recruitment and exaggerate the inflammatory response. The release of reactive oxygen species, granule contents, and neutrophil extracellular traps cause bystander damage to host cells. The migration of neutrophils across the endothelium, and in particular the epithelium, augments tissue damage. Disruption of the endothelial-epithelial barrier allows protein-rich fluid to enter the alveolar space, eventually resulting in alveolar flooding and respiratory failure. Alveolar Mφ, alveolar macrophage; NETosis, the process of cell death involving neutrophil extracellular trap (NET) formation.

Whereas ROS are generated in neutrophils by NADPH oxidase and nitric oxide synthase pathways, many soluble factors are prestored in neutrophil granules, the contents of which are released following transmigration and activation of neutrophils within the lung. Inhibiting the release of neutrophil granule contents has been shown to reduce lung injury and vascular permeability following challenge with the Streptococcus pyogenes M1 protein (159), further illustrating the contribution that neutrophils can make in promoting lung injury. An important proteinase released from neutrophil granules is neutrophil elastase, which is also elevated in human ARDS samples (43). The inhibition of neutrophil elastase has been demonstrated to reduce epithelial injury in several animal models, including a rat model of cystic fibrosis, a mouse model of pulmonary fibrosis, LPS-induced lung injury, mechanical ventilation-induced lung injury, and in colonic epithelial cells in vitro (56, 60, 74, 88, 170), although mice deficient in neutrophil elastase also have impaired host defense against gram-negative bacteria (14). The mechanisms by which neutrophil elastase causes lung injury are contentious, because it is unclear whether this proteinase directly damages endothelial or epithelial cells or whether tissue damage is the result of degradation of the alveolar basement membrane (26, 60). The use of neutrophil elastase inhibitors for the treatment of ARDS has therefore received much attention. For instance, sivelestat is routinely used in ARDS patients in Japan. However, a recent review of the available clinical data suggests that neutrophil elastase inhibition has no effect on mortality (81).

Other neutrophil-derived proteinases may also contribute to lung injury, namely proteinase-3, cathepsin-G, and several matrix metalloproteinases (MMPs). However, the nonspecific nature of their proteolytic activity means that multiple downstream effects can occur, other than extracellular matrix degradation. Proteinases have been shown to be capable of both activating and inactivating proinflammatory cytokines and chemokines, which is of particular relevance to ARDS. For example, MMP-9 increases the activity of IL-8 (CXCL8) through amino terminal processing but degrades CXCL1 (Gro-α) (175). The activation of chemokines enhances neutrophil migration, which may augment lung injury (160, 165), whereas the inactivation of proinflammatory cytokines and chemokines may be beneficial for the resolution of ongoing neutrophilia and lung inflammation (64, 119). MMP-9 endopeptidase activity also results in the generation of the extracellular matrix breakdown product of collagen, N-acetyl Pro-Gly-Pro (Ac-PGP), which is highly chemotactic for neutrophils and has been associated with inflammatory lung disease (57, 179). Ac-PGP can signal through the chemokine receptors CXCR1 and CXCR2 expressed by neutrophils, which in turn release more MMP-9 and thereby initiate a self-perpetuating cycle of neutrophilic inflammation (184). As a consequence of these multiple biological effects, it has been difficult to experimentally examine the relative importance of each proteinase during disease progression. Targeting neutrophil proteinases in the ARDS disease setting may also be impractical because of their numerous enzymatic targets.

Another important component of innate host defense is the release of NETs, through a process of cell death known as NETosis (19). NET formation involves the disintegration of the nuclear membrane, chromatin condensation, and the release of DNA and granule proteins into the extracellular space (20). NETosis can be initiated by various viral, bacterial, or fungal components and by host factors such as granulocyte-macrophage colony-stimulating factor, HMGB1, and activated platelets (29, 167). NET formation is also dependent on oxidant production (139), a deficiency that has been shown to reduce pathogen clearance from the lungs of infected mice in a chlorine gas model of lung injury (59). Although NETs have potent antimicrobial properties, they contain histones, enzymes, and peptides that are directly toxic to host cells. For example, inhibition of histones reduced mortality in mouse models of LPS-, TNF-, and cecal ligation-induced sepsis, whereas extracellular histones directly caused alveolar damage and pulmonary hemorrhage (183). Following infection with influenza virus H1N1 NETs were found to be associated with damaged alveoli in the lungs of challenged mice, whereas neutrophils released NETs onto the surface of influenza-infected epithelial cells in vitro and caused cell damage (133). The release of NETs has also been associated with lung injury following LPS, bacterial and fungal challenge (44, 174). Indeed, Toll-like receptor engagement on neutrophils is a potent activator of NET formation. NETs have also been observed in sterile transfusion-related acute lung injury (TRALI) in both mouse models and in human patients (168). Inhibiting extracellular histones or using DNaseI to disrupt NETs protected against TRALI (27). Furthermore, the lack of NET clearance from inflamed areas of the lung may further potentiate disease pathogenesis (86). Inhibiting NET formation, or key components of NETs, may therefore provide potential targets for the treatment of ARDS.

Neutrophil Heterogeneity

Rather than being short-lived, end-stage cells, increasing evidence suggests that different neutrophil subsets may be able perform distinct functions during inflammation and have the capacity to interact with various cells of the innate and adaptive immune systems (91). For example, it has been known for some time that the expression of CD16 (FcγRIII) varies on healthy human neutrophils (93). Those neutrophils that express abundant levels of CD16 (∼80–85% of blood neutrophils) are highly chemotactic to N-formyl-peptides, whereas those that weakly express CD16 (∼15–20%) do not respond. Three neutrophil subpopulations have been separated based on the expression of CD16 and CD62L (L-selectin): conventional (mature) neutrophils (CD16bright/CD62Lbright), banded (immature) neutrophils (CD16dim/CD62Lhigh), and a population of regulatory neutrophils (CD16bright/CD62Ldim) (84), the latter of which have reduced endothelial adhesion and reduced chemotactic capacity. These regulatory neutrophils are also able to modulate T cell proliferative responses via an interaction with the integrin αMβ2 (145). Furthermore, irrespective of underlying inflammation, neutrophils assume an activated phenotype (CD16bright/CD62Ldim/CD11bbright) after they migrate into the lung (50), suggesting that neutrophil migration across pulmonary endothelium and/or epithelium involves distinct molecular interactions that may be exclusive to the lung but independent of inflammation.

Neutrophil functional heterogeneity was further illustrated in an infection model, whereby neutrophils isolated from mice that were resistant to methicillin-resistant Staphylococcus aureus had a proinflammatory phenotype, whereas neutrophils from susceptible mice had an anti-inflammatory phenotype (173). Whereas proinflammatory neutrophils expressed IL-12 and CCL3 (MIP-1α), anti-inflammatory neutrophils expressed IL-10 and CCL2 (MCP-1). These phenotypically divergent neutrophils could also induce alternative macrophage populations (M1 vs. M2) (173), thereby modulating inflammatory vs. anti-inflammatory responses, respectively. Furthermore, in a human model of LPS-induced endotoxemia the circulating neutrophil compartment exhibited considerable functional heterogeneity (146). Following systemic inflammation, activated neutrophils (CD16bright) exhibited less chemotactic activity and increased ROS production compared with immature neutrophils (CD16dim) recently released from the bone marrow. It has also been suggested that the immunosuppression observed in the latter stages of systemic inflammatory response syndrome may be attributed to an increase in immature CD16dim neutrophils that have an unreactive phenotype, whereas reactive CD16bright neutrophils contribute to the initial phase of tissue damage (16, 145). This may suggest that inflammation results in a population of mature neutrophils that are capable of causing tissue damage, whereas immunosuppression is associated with an immature population that is more refractory to inflammatory signals (146). Neutrophils isolated from sepsis patients also have reduced CXCR2 expression and a corresponding loss of chemotactic activity (40), produce less ROS (5), and secrete the immunomodulatory cytokine IL-10 (87). These factors may contribute to the immunosuppressive state in progressive sepsis and account for the increased susceptibility to bacterial pneumonia. However, it is still uncertain whether divergent subsets of neutrophils represent distinct cell lineages (33) or whether they just reflect functional heterogeneity (16) because of their inherent phenotypic plasticity (82).

It has recently been demonstrated that neutrophils are capable of undergoing retrograde transendothelial migration to reenter the circulation (21). In so doing, they undergo a phenotypic change characteristic of inflammatory neutrophils (ICAM-1high/CXCR1low) that are primed for ROS production. These neutrophils may also contribute to the dissemination of systemic inflammation, as ICAM-1high neutrophils that were generated following ischemia-reperfusion injury were sequestered to the pulmonary microvasculature and caused lung inflammation and increased endothelial-epithelial barrier permeability (182). Neutrophils that have undergone retrograde transendothelial migration may account for the activated subsets (CD16bright/CD62Ldim) found in the circulation following inflammation (146) and may therefore contribute to the pathogenic state of inflammatory lung diseases. Targeting specific neutrophil subpopulations may be a useful approach for modulating the inflammation associated with ARDS.

Mechanisms of Neutrophil Recruitment into the Lung

Numerous factors have been associated with neutrophil recruitment into the lung and alveolar spaces in animal models of ARDS, including cell adhesion molecules, proteinases, oxidants, cytokines, and chemokines. The substantial pulmonary vasculature, and in particular the capillary bed located within the distal lung, contribute to making this organ a unique site for neutrophil egress. In most organs, neutrophils gain access to tissue compartments by migrating across high endothelial venules (HEVs), which are associated with high flow rates and sheer forces. However, in the distal lung, neutrophils migrate across narrow pulmonary capillaries, which have low tidal forces and diameters smaller than the neutrophils themselves [≥2 μm compared with 6–8 μm (22)]. These unique migratory parameters lengthen the time available for transmigration and allow neutrophils to extravasate without the need for the conventional rolling and adhesion processes that occur in HEVs. For example, neither L-selectin nor β2-integrins are essential for neutrophil migration across alveolar capillaries (97, 128). Furthermore, it has been estimated that neutrophils sequestered within the pulmonary microvasculature account for a large pool of neutrophils that likely exceeds the size of the circulating pool by as much as fivefold (42). Such a large marginated pool of neutrophils within the lung microvasculature makes them ideally located to respond immediately to an inflammatory insult. Indeed, neutrophils can rapidly cross the endothelial cell wall in as little as 2 min, preferentially at the corner junctions where three endothelial cells connect (23). Migration at endothelial tricell corners is thought to avoid disrupting intercellular tight junctions, thereby maintaining normal capillary barrier function. Considering that the lungs are in continuous contact with environmental insults, the transmigration across the endothelium and the activation of neutrophils within this unique organ are tightly regulated.

In the context of the lung microenvironment, neutrophils must undergo extensive shape change to migrate across both the capillary endothelium and alveolar epithelium. Neutrophil extravasation is also likely to involve the release of proteinases and elastases (90) that allow movement through the basement membrane by degrading the extracellular matrix, although the precise role of specific proteinases, such as elastase and gelatinase, remains controversial (22, 76). Migration of neutrophils out of the endothelium and across the pulmonary interstitium is thought to be mediated by fibroblasts, which establish defined conduits that assist neutrophils to gain access to the basal surface of the epithelium (177), while at the same time maintaining capillary-alveolar integrity (157). These conduits are likely to be anatomically different to the interalveolar connections, known as the pores of Kohn, which connect adjoining alveolar sacs (37). These interalveolar connections are the size of an alveolar epithelial cell and act to maintain gaseous exchange throughout the respiratory cycle. Although alveolar macrophages are known to migrate between adjacent alveoli through pores of Kohn (142), there is no evidence that neutrophils use these structures during transepithelial migration.

The migration of neutrophils across the alveolar epithelium is also thought to involve several key steps (192) including adhesion, migration, and postmigration events. Adhesion of primed neutrophils to the basolateral surface of the epithelium likely involves the binding of β2-integrins (CD11b/CD18) on the surface of neutrophils to ICAM-1 on the epithelium (10). Furthermore, ICAM-1 is upregulated on alveolar epithelial cell surfaces in response to several inflammatory stimuli (171, 172). Migration of neutrophils across the epithelium is thought to occur between adjacent epithelial cells through the paracellular space, although it is uncertain whether the mere mechanics of this process is sufficient to increase alveolar permeability. In studies using intestinal epithelial cells, for example, neutrophil migration causes the loss of epithelial tight junctions without any apparent disruption in epithelial permeability (125, 134). Neutrophils are also thought to utilize specific epithelial invasion sites, at least when migrating across the intestinal epithelium (134, 140), which may lessen barrier disruption. Alveolar epithelial cell injury was shown to be dependent on the activation of neutrophils and the release of proteinases and elastase, although independent of ROS generation or direct cytolysis (7, 156). Zemans et al. (191) demonstrated that neutrophil migration across human epithelial type II cells causes disruption of the monolayer, quickly followed by reepithelialization, a process that was dependent on β-catenin signaling (193). Therefore, this would suggest that excessive neutrophil migration across the alveolar epithelium, and the associated release of degrading enzymes, overwhelms normal physiological migration pathways and directly contributes to epithelial damage and permeability within the inflamed lung. The ability of the pulmonary epithelium to repair itself after injury may be a critical factor in determining the severity of disease (39).

Postmigration events involve the adhesion of neutrophils to the apical surface of epithelial cells within the alveolar lumen. However, it is not clear what molecular interactions are involved in this process. One possibility is an interaction between neutrophil selectins and epithelial ICAM-1 (10, 131, 132), since this interaction clearly influences neutrophil migration into the air space. However, the role that this interaction plays in alveolar neutrophil margination remains to be confirmed. Another possibility is that neutrophil Fc receptors bind to soluble antibody attached to the apical surface of epithelial cells, which has been demonstrated on the intestinal epithelium in certain gut pathologies (148). Furthermore, ARDS has been associated with a deficiency in the clearance of neutrophils from the alveolar space, which may be directly related to a defect in neutrophil apoptosis and a corresponding prolonged lifespan (117). Indeed, several cytokines and growth factors such as granulocyte colony-stimulating factor (G-CSF) and IL-8, which are elevated in ARDS, have prosurvival effects on neutrophils (3). A decrease in neutrophil apoptosis, combined with a deficiency in cell clearance, may also contribute to disease pathogenesis (106).

Role of Chemokines in Neutrophil Recruitment to the Lung

The chemokine IL-8, otherwise known as CXCL8, and the rodent homologues CXCL1 (KC) and CXCL2 (MIP-2) are thought to be central to neutrophil recruitment into the lung during ARDS. Important correlations have been made from clinical ARDS samples, including pulmonary edema aspirates and BALF, between increased IL-8 concentrations, disease severity (65, 126), and neutrophil migration into air spaces (127). These studies provide important information regarding the role that neutrophils and IL-8 play in the pathogenesis of ARDS. Although the related CXC chemokines CXCL5 (ENA-78) and CXCL1 (Gro-α) are also elevated in BALF from ARDS patients (63), only IL-8 consistently correlates with the number of neutrophils and disease severity (61, 176). Furthermore, IL-8 has been demonstrated to be the most potent neutrophil chemoattractant in BALF from ARDS patients and was the predominant neutrophilic chemokine released from LPS-stimulated human alveolar macrophages (62, 101, 127). Therefore IL-8 is considered the main neutrophil chemoattractant in ARDS. The levels of IL-8 in ARDS BALF also correlate with survival, as IL-8 levels are lower in survivors compared with nonsurvivors (12). Neutrophil activation also directly correlates with disease severity and with the levels of IL-8, TNF, and IL-6 (31). Another study revealed that IL-8, as well as G-CSF, correlates with neutrophil influx and the severity of ARDS (3). Wiedermann et al. (180) further demonstrated that IL-8 correlates with neutrophil numbers in BALF of ARDS patients, and G-CSF with circulating neutrophil numbers, whereas CXCL5 (ENA-78) did not show any correlation (Table 1).

Table 1.

Chemokines in ARDS — clinical studies

Chemokine Disease Effect Reference
CXCL1 ARDS ↑ in BALF. Levels consistent with neutrophil numbers. Poor neutrophil chemokine in ARDS BALF. 51, 71, 176
(Gro-α)
In vitro Induced neutrophil chemotaxis, shape change and activation 51
CXCL2 Sepsis Polymorphism associated with higher rate of mortality. 109
(Gro-β)
In vitro Induced neutrophil chemotaxis, shape change and activation 51
CXCL4 ARDS ↑ in BALF in association with CCL5-CXCL4 heteromers. Possible associated with neutrophil influx. 66
(PF-4)
CXCL5 ARDS ↑ in BALF. Levels inconsistent with neutrophil numbers. Weak neutrophil chemokine in ARDS BALF. 51, 63, 180
(ENA-78)
CXCL8 ARDS ↑ in BALF and pulmonary edema fluid. Levels correlate with neutrophil numbers. Levels correlate with disease severity. Levels correlate with outcome. Predominant neutrophil chemokine in ARDS BALF. 3, 12, 31, 32, 61, 65, 107, 126, 176
(IL-8)
In vitro Removal of IL-8 or use of anti-IL-8 antibody blocks neutrophil activation and chemotaxis. Chemotaxis not completely removed however. 52, 71, 127
CCL2 ARDS ↑ in BALF. Levels correlate with lung injury. 63
(MCP-1)
CCL3 ARDS ↑ in BALF. No correlation with neutrophil number. 63
(MIP-1α)
CCL4 LPS ex vivo lung ↑ in BALF. Increased levels coincided with increased neutrophils. (IL-8, CXCL5, CCL3, and CCL2 also upregulated). 135
(MIP-1β)
CCL5 ARDS ↑ in BALF in association with CCL5-CXCL4 heteromers. Possible association with neutrophil influx. 66
(RANTES)
COPD ↑ in bronchial biopsies. Associated with increased activation and upregulation of CC-chemokine receptors on neutrophils. 38, 70

ARDS, acute respiratory distress syndrome; Gro-α/β, growth stimulating activity-α/β; BALF, bronchoalveolar lavage fluid; PF-4, platelet factor-4; ENA-78, epithelial cell-derived neutrophil-activating peptide-78; IL-8, interleukin-8; MCP-1, monocyte chemoattractant protein-1; MIP-1α/β, macrophage inflammatory protein-1α/β; RANTES, regulated on activation, normal T cell expressed and secreted; LPS, lipopolysaccharide; COPD, chronic obstructive pulmonary disease.

IL-8 is able to bind to either of two receptors, CXCR1 or CXCR2, on the surface of human neutrophils, although the relative contribution of each receptor to disease pathogenesis is still uncertain. Until recently, CXCR2 was thought to be the only functional homologue in rodents, whereas extensive neutralization and antagonism studies clearly demonstrated its importance for neutrophil migration into the inflamed lung (92). A functional CXCR1 homologue has now been discovered in mice, although its importance in neutrophil migration is not clear (129). In humans, both receptors are internalized following ligation with IL-8 but only CXCR1 is rapidly reexpressed on the cell surface (34). It has also been demonstrated that IL-8 stimulation of CXCR2 acts to prime neutrophils for CXCR1 receptor activation, as measured by Ca2+ mobilization (71). CXCR1 is therefore thought to be the major receptor for IL-8 on human neutrophils. Continued ligation of CXCR1 with IL-8 or CXCL1 is also able to prevent neutrophil apoptosis and may therefore be involved in the abnormal clearance of neutrophils in ARDS (45).

Numerous animal studies, notably using the LPS model of lung inflammation or the acid aspiration-induced lung injury model, clearly demonstrate the central role of IL-8 homologues in driving neutrophil influx into the inflamed lung (67). For example, administration of an anti-IL-8 antibody prevented neutrophilic lung inflammation and extravascular leak in an acid-aspiration rabbit model (49) and an LPS-induced rabbit model of lung injury (189). The importance of IL-8 in recruiting neutrophils to sites of inflammation has also been demonstrated in several nonpulmonary disease models, as anti-IL-8 antibody treatment was able to prevent neutrophil-mediated tissue damage in rabbit models of LPS-induced dermatitis, LPS/IL-1-induced arthritis, lung reperfusion injury, and acute immune complex-type glomerulonephritis (69). IL-8 is therefore considered to be the archetypal neutrophil chemoattractant.

Mouse CXCL1 (KC), the functional homologue of human IL-8, also binds to the receptors CXCR1 and CXCR2. However, the chemokine network is considered to possess a high degree of redundancy and pleiotropy, as the related ELR+ chemokines CXCL2 (MIP-2α), CXCL3 (MIP-2β), CXCL5 (ENA-78), and CXCL15 (lungkine) can also bind CXCR1 and CXCR2. However, these chemokines have varying levels of neutrophil chemotactic potency in humans, with CXCL8 being the most potent followed by CXCL1, CXCL5, CXCL2, and CXCL3, in that order (52). As well as inducing varying levels of chemotaxis, these chemokines also have differential effects on neutrophil activation, exocytosis, and respiratory burst (58), indicating that related CXC-chemokines have different functional activities on neutrophils despite sharing the same receptors (Table 2). In addition, these chemokines may be able to functionally compensate for the absence of CXCL1 (or IL-8), suggesting that therapeutically targeting a single ELR+ chemokine may not be an appropriate approach in ARDS. Neutralizing CXCL1 in the bronchoalveolar lavage fluid of LPS-treated rats also reduced neutrophil migration but only by ∼50% (53), suggesting that other neutrophil chemotactic factors may be present in the lavage fluid from the inflamed lung. For example, CXCL2 has also been shown to regulate neutrophil migration and function in a mouse model of LPS-induced (6) and sepsis-induced lung injury (108). CXCL1 and CXCL2 both signal through CXCR2 and, although they have broadly similar effects on neutrophil recruitment, they alter neutrophil function in slightly different ways (110). The local distribution of CXCL1 and CXCL2 within the lung may be important in determining the extent of neutrophil migration, activation, and subsequent tissue injury. Antagonism of CXCR2 is therefore felt to provide a more suitable approach than targeting individual chemokines, as this would inhibit the signaling and chemotactic potential of several chemokines, in particular CXCL1 and CXCL2, and of the PGP tripeptide (158). Indeed, mice deficient in CXCR2 (CXCR2−/−) have been shown to have diminished neutrophil recruitment into the lung in response to LPS (150). Neutrophil migration out of the bone marrow (46) and across the pulmonary endothelium following lung injury has also been shown to be regulated by CXCR2 (92, 164).

Table 2.

Chemokines in ARDS — animal studies

Chemokine Model of Lung Injury Animal Effect Reference
CXCL1 LPS-induced Mouse ↑ in BALF. Neutralization decreased neutrophil recruitment. 6, 190
(KC/CINC)
Endotoxemia/LPS-induced Rat ↑ in BALF. Neutralization decreased neutrophil recruitment. Recombinant CXCL1 increased neutrophil influx. 25, 53, 187
Acute pancreatitis-induced lung injury Rat Neutralization decreased lung permeability and MPO activity. 17
Ventilator-induced lung injury Mouse ↑ in BALF. CXCR2−/− mice had reduced neutrophil sequestration and lung injury. 15
Klebsiella infection Mouse CXCL1 mediated upregulation of CXCL2/CXCL5. Host defense dependent on CXCL1. Required for ROS generation in neutrophils. 11, 24
CXCL2 LPS-induced Mouse ↑ in BALF. Endogenous DARC sequestration or syndecan-1 shedding decreased neutrophil recruitment. 6, 72, 149
(MIP-2)
LPS-induced Rat ↑ in BALF. Neutralization diminished neutrophil accumulation. Released by alveolar macrophages and epithelial cells. 68, 112, 152, 155
Endotoxemia Mouse Neutralization decreased neutrophil recruitment into lung. Recued lung permeability. 2, 161
Acid-induced Rat ↑ in BALF. Neutralization decreased neutrophil recruitment, leak and improved oxygenation. 154
Ventilator-induced lung injury Mouse ↑ in BALF. Increased levels correlated with neutrophil accumulation and lung injury. 15
Ventilator-induced lung injury Rat ↑ in BALF. Neutralization decreased neutrophil recruitment and leak. 147
CXCL4 LPS-, sepsis-, and acid-induced Mouse Neutralization abolished neutrophil recruitment, permeability and lung damage. CCL5-CXCL4 formed heterodimers. 66
(PF-4)
CXCL5 LPS-induced Mouse CXCL5 dominated neutrophil recruitment into the lung over CXCL1 and CXCL2. 121
(ENA-78)
Escherichia coli infection Mouse CXCL5−/− mice had impaired neutrophil influx into the lung. 121, 122
CXCL8 LPS-induced Rabbit ↑ in BALF. Neutralization decreases neutrophil recruitment, permeability and gas exchange. 69, 186, 189
(IL-8)
LPS/oleic acid double hit Rabbit ↑ in BALF. Anti-IL-8 neutralization blocks neutrophil infiltration. 9
Lung reperfusion injury Rabbit ↑ in BALF. Neutralizations prevented neutrophil infiltration and tissue injury. 153
Acid-aspirate injury Rabbit ↑ in BALF. Neutralization decreased neutrophil accumulation, leak, and restored lung function and oxygenation. Neutralization improved fluid clearance. 49
CXCL10 Oxidative stress Mouse ↑ in BALF. Neutralization decreases neutrophil accumulation. 124
(IP-10)
CXCL12 LPS-induced Mouse Chemoattractant and anti-apoptotic mediator for lung neutrophils ex vivo. 185
(SDF-1)
CCL2 LPS-induced Mouse ↑ in lung. Neutralization decreased neutrophil recruitment. 83, 123
(MCP-1)
Endotoxemia Mouse ↑ in plasma and lung. Anti-CCL2 resulted in increased mortality and TNF levels. 194
E. coli infection Mouse CCL2−/− mice had reduced bacterial clearance. Recombinant CCL2 induced neutrophil influx. Neutrophils responded in CCL2 In vitro. 8
IgG immune complex Rat Neutralization had no effect on neutrophil recruitment. 18
CCL3 Trauma-hemorrhage Mouse ↑ in serum. CCL3−/− mice had decreased edema and lung MPO activity. 79
(MIP-1α)
Lung reperfusion injury Rat ↑ in lungs. Neutralization decreased neutrophil infiltration. 94
CCL4 IgG immune complex Rat Neutralization decreased neutrophil recruitment and vascular leak. 18
(MIP-1β)
CCL5 LPS-, sepsis-, and acid-induced Mouse ↑ in BALF. Neutralization abolished neutrophil recruitment, permeability and lung damage. CCL5-CXCL4 heterodimers may be involved. 66
(RANTES)
IgG immune complex Rat No change in expression. Inhibition had no effect on neutrophil recruitment. 18
CCL7 Oxidative stress Mouse Neutralization decreased neutrophil recruitment. 124
(MCP-3)
LPS-induced Mouse ↑ in lung. Neutralization decreased neutrophils recruitment into air space. 123

KC, keratinocyte-derived chemoattractant; CINC, cytokine-induced neutrophil chemoattractant; ROS, reactive oxygen species; DARC, Duffy antigen receptor for chemokines; IL-8, interleukin-8; IP-10, interferon γ-induced protein 10; SDF-1, stromal cell-derived factor-1; MCP-1/3, monocyte chemoattractant protein-1/3; IgG, immunoglobulin-G.

Extrapolating information from studies performed in animal models of lung injury call for caution, however, because of the differences between mouse and human chemokine systems. For example, CXCL5 does not consistently correlate with human disease severity, despite mouse models of ARDS clearly demonstrating an important role for this chemokine in neutrophil recruitment (180). Furthermore, CXCR1 acts only as a weak receptor for neutrophil chemoattraction in mice, since CXCR2 seems to be more important, whereas in humans CXCR1 appears to be the dominant neutrophil receptor. Neutrophils isolated from the blood of sepsis patients also express lower levels of CXCR2 compared with neutrophils from healthy individuals (40), whereas IL-8 and CXCL1 both downregulate CXCR2 upon stimulation (45). Another caveat is that CXCR2 neutralization may reduce the effectiveness of innate immune defenses and consequently result in uncontrolled bacterial or fungal infection (73, 120). It will therefore be important to consider the appropriateness of targeting CXCR2, or indeed the neutrophilic innate immune response, in certain patient groups with an underlying infection. Despite these species differences, and the potential for microbial outgrowth, a number of promising CXCR2 antagonists are being developed for use in humans to treat neutrophilic lung disease (28, 78, 92, 102).

IL-8 Immune Complexes in ARDS

A significant fraction of the IL-8 recovered from the BALF of ARDS patients is complexed to endogenous IgG, mainly of the IgG3 and IgG4 subclasses (98, 166). Furthermore, elevated levels of IL-8 immune complexes have been associated with poor clinical outcome in ARDS patients and in patients who are at risk of developing ARDS (98100). IL-8 immune complexes may therefore provide a useful biomarker in ARDS (55). It has been shown that these anti-IL-8:IL-8 complexes retain both their chemotactic properties for neutrophils and their ability to activate them, resulting in the release of oxidants and MPO, a function that is dependent on the Fc receptor, FcγRIIa (96). Furthermore, anti-IL-8:IL-8 complexes inhibit neutrophil apoptosis, which is associated with an increase in the expression of Bcl-xL and a decrease in Bak and Bax (54). Indeed, it is well established that apoptosis of neutrophils is delayed in patients with ARDS (103). In addition, these IL-8 immune complexes activate endothelial cells, which upregulate ICAM-1 and increase ERK, JNK, and Akt phosphorylation (95), which may suggest that IL-8 immune complexes can induce endothelial cells to release proinflammatory mediators. Indeed, FcγRIIa is expressed on several cell types in the ARDS lung, including endothelial cells, neutrophils, and cells of the myeloid lineage, such as macrophages, monocytes, and dendritic cells (4).

The significance of these IL-8 immune complexes remains uncertain, however. The original paper by Kurdowska et al. (98) suggests that each complex comprises just one IL-8 molecule and one IgG molecule. Even though these immune complexes colocalize with FcγRIIa in ARDS lungs (4), it is unclear how these anti-IL-8:IL-8 complexes signal via FcγRIIa. For example, cross-linking is normally required for Fc receptor activation, unless these antibodies are tethered to a cell surface. This is in contrast to the proinflammatory and prochemotactic activity of IL-8 antibody complexes that are dependent on FcγRIIa (96). The other explanation is that these complexes represent a means of regulating the IL-8 response by sequestering the chemokine and preventing functional activity (98), although later studies suggest that these complexes are inflammatory, not regulatory (96). The situation may be further complicated by the expression of FcγRIIb within the lung, which is a known inhibitory receptor that may have opposing downstream effects to FcγRIIa (35). Another question arises: are these autoantibodies produced by everybody or are they limited to patients with ARDS? Are these autoantibodies produced by such individuals prior to ARDS or are they generated in response to ongoing inflammation? Furthermore, IgG antibody isotypes are normally located in the circulation rather than in the lumen of mucosal tissues. Considering these IL-8 immune complexes are readily recovered from BALF, it is therefore unclear whether they are generated locally or whether they enter the alveolar space as a consequence of microvascular leak. Finally, does ARDS therefore have an autoimmune component? This is unlikely, as ARDS is neither a spontaneous disease of endogenous origin nor a chronic inflammatory disease.

Other Chemokines Involved in Neutrophil Migration

It has been demonstrated that human neutrophils alter their cell surface expression of several chemokine receptors in the lung during episodes of inflammation, which may have important implications for ARDS pathogenesis. For example, Hartl et al. (70) demonstrated that human neutrophils isolated from the BALF of chronic obstructive pulmonary disease (COPD) patients expressed higher levels of CCR1, CCR2, CCR3, CCR5, CXCR3, and CXCR4 compared with circulating neutrophils (they found similar upregulation on neutrophils from the synovium of arthritis patients). The same study showed that neutrophils isolated from COPD BALF were able to chemotax toward CCL2 (MCP-1), CCL3 (MIP-1α), CCL4 (MIP-1β), CCL11 (eotaxin), CXCL11 (I-TAC), and CXCL12 (SDF-1). Although these samples were obtained from COPD and not ARDS patients, this does suggest that neutrophils are potentially capable of responding to a number of CC and CXC chemokines other than IL-8. Some of these chemokines, CXCL11 and CCL5 for example, are also elevated in the BALF of COPD patients (38). CCL2 and CCL3 levels were also demonstrated to be consistently elevated in the BALF of ARDS patients, although they did not correlate with either neutrophil or macrophage numbers (63). In addition, it has been reported that human neutrophils downregulate CXCR1 and CXCR2 after recruitment to the airways (144), which may have important consequences for the therapeutic inhibition of these receptors in lung disease. Indeed, human neutrophils have been shown to undergo rapid gene expression changes following transepithelial migration, resulting in delayed apoptosis and reduced capacity for IL-8-mediated chemotaxis (36).

The platelet-derived chemokines CCL5 and CXCL4 are also elevated in human ARDS samples and mouse models of lung injury. These chemokines have been shown to form heteromers and their expression correlates with neutrophil efflux into mouse lung tissue (66). Disruption of these heteromers attenuated experimental sepsis and LPS-induced lung injury. LPS-treated mice also have elevated levels of the related chemokine CXCL12 and increased migration of CXCR4+ neutrophils (185). Moreover, CXCL12 was shown to reduce neutrophil apoptosis in inflamed lungs. Struyf et al. (163) further demonstrated that CXCL6 can synergize with CCL7 (MCP-3) to promote neutrophil migration, albeit in a mouse model of peritoneal inflammation. CCL7, together with CXCL10, was also shown to orchestrate neutrophil migration into the lung in response to oxidative stress (124), further suggesting that neutrophil responses are influenced by a variety of mediators in certain inflammatory conditions. CXCL10 has also been implicated in acid-induced lung injury and ARDS resulting from influenza infection of mice (80). Neutrophils that expressed CXCL10 were able to signal in an autocrine manner via CXCR3, which enhanced neutrophil oxidative burst, chemotaxis, and lung inflammation. Furthermore, CXCR3 was specifically expressed on neutrophils that infiltrated the lung, suggesting that this subset of neutrophils may provide a suitable target for the treatment of ARDS (80).

In a mouse model of Escherichia coli-induced lung injury, MCP-1−/− mice showed reduced CCR2+ neutrophil recruitment into the lungs (8). CCR1−/− and CCR2−/− mice also demonstrated reduced neutrophilic inflammation following LPS challenge, although no differences were observed for CCR3−/− mice (188). In contrast, inhibition of CCR3 expressed on pulmonary epithelial cells reduced IL-8 secretion and neutrophil recruitment in response to LPS (105). CCR1 and CCR2 are also upregulated on neutrophils isolated from adjuvant-challenged rats, allowing them to respond CCL2 (MCP-1) in in vitro chemotaxis assays (82). However, neutrophil migration into the lung has been shown to be independent of CCR2 (and CCL2) but cooperatively dependent on CCR2+ monocyte recruitment (118). Likewise in an IgG immune complex-induced rat model of lung injury, neutralization of CCL2 had no effect on neutrophilic inflammation, although neutralization of CCL4 (MIP-1β) did reduce vascular permeability, neutrophil recruitment, and TNF production in BALF (18).

More recently, we have demonstrated that neutralizing CCL2, or the related chemokine CCL7, significantly diminishes neutrophil accumulation in the air spaces of LPS-challenged mice (123). Furthermore, neutrophils isolated from inflamed lungs displayed decreased expression of cell surface CXCR2 and increased expression of CCR1 and CCR2 (but not CCR3). It is therefore becoming increasingly clear that the neutrophilic chemokine network within the lung is more complex than previously thought and may be dependent on the contextual influence of several inflammatory and chemotactic factors. It is also likely that the migration of neutrophils into various tissue microcompartments, for example endothelial vs. epithelial, is regulated by distinct sets of chemotactic cues differentially expressed within certain lung microenvironments (123), suggesting there may exist a significant amount of nonredundancy within the lung chemokine network (Tables 1 and 2). The functional capacity of individual chemokines may need to be considered within the context of the surrounding inflammatory milieu and the tissue compartment in which they operate.

SUMMARY

Neutrophils play an important role in innate immune defense by phagocytosing pathogenic bacteria and preventing further invasion through the release of ROS, proteinases, antimicrobial peptides, and NETs. However, the excessive accumulation of activated neutrophils can cause unwanted bystander tissue damage, which can negatively affect the normal physiology of the lung by modulating endothelial-epithelial barrier integrity, resulting in pulmonary edema and hypoxemia (Fig. 1). The migration of large numbers of neutrophils across the alveolar epithelium may significantly contribute to epithelial permeability. In addition, proteinases such as neutrophil elastase and matrix metalloproteinases directly degrade the extracellular matrix and components of the paracellular space. The excessive release of reactive mediators, such as oxidants, cationic antimicrobial peptides, and lipid mediators, by neutrophils attached to the apical surface of the alveolar epithelium, may also cause damage and contribute to endothelial-epithelial barrier permeability. Capillary-alveolar permeability is a major hallmark of ARDS, resulting in alveolar leak, lung edema, deficient oxygenation, and hypoxemia. Therefore, targeting excessive neutrophil accumulation within this disease setting may represent a viable approach for the treatment of ARDS.

Studies performed with clinical ARDS samples and animal models of ARDS have revealed a central role for IL-8 (CXCL8) in regulating neutrophil recruitment into the lungs, and consequent tissue damage, capillary-alveolar permeability, and oxygen deficiency. However, the chemokine network regulating neutrophil migration in the lung may involve multiple chemokine ligand families, including several CC-chemokines such as CCL2, CCL4, and CCL7, thereby expanding the number of potential therapeutic targets in this disease setting. The mechanism of neutrophil recruitment into the lung is a multistep process involving extravasation across the pulmonary endothelium, trafficking across the lung interstitium, and migration across the alveolar epithelium into air spaces. This involves multiple chemotactic molecules acting at each of the spatial-temporal checkpoints. Further research needs to be undertaken to better understand these complex chemokine networks and the contribution of individual chemokines during the development of both pulmonary and nonpulmonary diseases associated with neutrophilic inflammation.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

AUTHOR CONTRIBUTIONS

A.E.W. prepared figures; A.E.W. drafted manuscript; A.E.W. and R.C.C. edited and revised manuscript; A.E.W. and R.C.C. approved final version of manuscript.

ACKNOWLEDGMENTS

The authors are grateful to the Medical Research Council U.K. (grant no G0800265) and the Rosetrees Trust for funding support received for ARDS research within the Centre.

REFERENCES

  • 1.Abraham E. Neutrophils and acute lung injury. Crit Care Med 31: S195–S199, 2003 [DOI] [PubMed] [Google Scholar]
  • 2.Abraham E, Carmody A, Shenkar R, Arcaroli J. Neutrophils as early immunologic effectors in hemorrhage- or endotoxemia-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 279: L1137–L1145, 2000 [DOI] [PubMed] [Google Scholar]
  • 3.Aggarwal A, Baker CS, Evans TW, Haslam PL. G-CSF and IL-8 but not GM-CSF correlate with severity of pulmonary neutrophilia in acute respiratory distress syndrome. Eur Respir J 15: 895–901, 2000 [DOI] [PubMed] [Google Scholar]
  • 4.Allen TC, Fudala R, Nash SE, Kurdowska A. Anti-interleukin 8 autoantibody:interleukin 8 immune complexes visualized by laser confocal microscopy in injured lung. Arch Pathol Lab Med 131: 452–456, 2007 [DOI] [PubMed] [Google Scholar]
  • 5.Alves-Filho JC, Spiller F, Cunha FQ. Neutrophil paralysis in sepsis. Shock 34, Suppl 1: 15–21, 2010 [DOI] [PubMed] [Google Scholar]
  • 6.Aoki K, Ishida Y, Kikuta N, Kawai H, Kuroiwa M, Sato H. Role of CXC chemokines in the enhancement of LPS-induced neutrophil accumulation in the lung of mice by dexamethasone. Biochem Biophys Res Commun 294: 1101–1108, 2002 [DOI] [PubMed] [Google Scholar]
  • 7.Ayars GH, Altman LC, Rosen H, Doyle T. The injurious effect of neutrophils on pneumocytes in vitro. Am Rev Respir Dis 129: 964–973, 1984 [DOI] [PubMed] [Google Scholar]
  • 8.Balamayooran G, Batra S, Balamayooran T, Cai S, Jeyaseelan S. Monocyte chemoattractant protein 1 regulates pulmonary host defense via neutrophil recruitment during Escherichia coli infection. Infect Immun 79: 2567–2577, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bao Z, Ye Q, Gong W, Xiang Y, Wan H. Humanized monoclonal antibody against the chemokine CXCL-8 (IL-8) effectively prevents acute lung injury. Int Immunopharmacol 10: 259–263, 2010 [DOI] [PubMed] [Google Scholar]
  • 10.Basit A, Reutershan J, Morris MA, Solga M, Rose CE, Jr, Ley K. ICAM-1 and LFA-1 play critical roles in LPS-induced neutrophil recruitment into the alveolar space. Am J Physiol Lung Cell Mol Physiol 291: L200–L207, 2006 [DOI] [PubMed] [Google Scholar]
  • 11.Batra S, Cai S, Balamayooran G, Jeyaseelan S. Intrapulmonary administration of leukotriene B(4) augments neutrophil accumulation and responses in the lung to Klebsiella infection in CXCL1 knockout mice. J Immunol 188: 3458–3468, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Baughman RP, Gunther KL, Rashkin MC, Keeton DA, Pattishall EN. Changes in the inflammatory response of the lung during acute respiratory distress syndrome: prognostic indicators. Am J Respir Crit Care Med 154: 76–81, 1996 [DOI] [PubMed] [Google Scholar]
  • 13.Bdeir K, Higazi AA, Kulikovskaya I, Christofidou-Solomidou M, Vinogradov SA, Allen TC, Idell S, Linzmeier R, Ganz T, Cines DB. Neutrophil alpha-defensins cause lung injury by disrupting the capillary-epithelial barrier. Am J Respir Crit Care Med 181: 935–946, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Belaaouaj A, McCarthy R, Baumann M, Gao Z, Ley TJ, Abraham SN, Shapiro SD. Mice lacking neutrophil elastase reveal impaired host defense against gram negative bacterial sepsis. Nat Med 4: 615–618, 1998 [DOI] [PubMed] [Google Scholar]
  • 15.Belperio JA, Keane MP, Burdick MD, Londhe V, Xue YY, Li K, Phillips RJ, Strieter RM. Critical role for CXCR2 and CXCR2 ligands during the pathogenesis of ventilator-induced lung injury. J Clin Invest 110: 1703–1716, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Beyrau M, Bodkin JV, Nourshargh S. Neutrophil heterogeneity in health and disease: a revitalized avenue in inflammation and immunity. Open Biol 2: 120–134, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bhatia M, Brady M, Zagorski J, Christmas SE, Campbell F, Neoptolemos JP, Slavin J. Treatment with neutralising antibody against cytokine induced neutrophil chemoattractant (CINC) protects rats against acute pancreatitis associated lung injury. Gut 47: 838–844, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bless NM, Huber-Lang M, Guo RF, Warner RL, Schmal H, Czermak BJ, Shanley TP, Crouch LD, Lentsch AB, Sarma V, Mulligan MS, Friedl HP, Ward PA. Role of CC chemokines (macrophage inflammatory protein-1 beta, monocyte chemoattractant protein-1, RANTES) in acute lung injury in rats. J Immunol 164: 2650–2659, 2000 [DOI] [PubMed] [Google Scholar]
  • 19.Brinkmann V, Laube B, Abu Abed U, Goosmann C, Zychlinsky A. Neutrophil extracellular traps: how to generate and visualize them. J Vis Exp 36: e1724, 2010. 10.3791/1724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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] [PubMed] [Google Scholar]
  • 21.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] [PubMed] [Google Scholar]
  • 22.Burns AR, Smith CW, Walker DC. Unique structural features that influence neutrophil emigration into the lung. Physiol Rev 83: 309–336, 2003 [DOI] [PubMed] [Google Scholar]
  • 23.Burns AR, Walker DC, Brown ES, Thurmon LT, Bowden RA, Keese CR, Simon SI, Entman ML, Smith CW. Neutrophil transendothelial migration is independent of tight junctions and occurs preferentially at tricellular corners. J Immunol 159: 2893–2903, 1997 [PubMed] [Google Scholar]
  • 24.Cai S, Batra S, Lira SA, Kolls JK, Jeyaseelan S. CXCL1 regulates pulmonary host defense to Klebsiella infection via CXCL2, CXCL5, NF-kappaB, and MAPKs. J Immunol 185: 6214–6225, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Calkins CM, Bensard DD, Shames BD, Pulido EJ, Abraham E, Fernandez N, Meng X, Dinarello CA, McIntyre RC., Jr IL-1 regulates in vivo C-X-C chemokine induction and neutrophil sequestration following endotoxemia. J Endotoxin Res 8: 59–67, 2002 [PubMed] [Google Scholar]
  • 26.Carden D, Xiao F, Moak C, Willis BH, Robinson-Jackson S, Alexander S. Neutrophil elastase promotes lung microvascular injury and proteolysis of endothelial cadherins. Am J Physiol Heart Circ Physiol 275: H385–H392, 1998 [DOI] [PubMed] [Google Scholar]
  • 27.Caudrillier A, Kessenbrock K, Gilliss BM, Nguyen JX, Marques MB, Monestier M, Toy P, Werb Z, Looney MR. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J Clin Invest 122: 2661–2671, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chapman RW, Minnicozzi M, Celly CS, Phillips JE, Kung TT, Hipkin RW, Fan X, Rindgen D, Deno G, Bond R, Gonsiorek W, Billah MM, Fine JS, Hey JA. A novel, orally active CXCR1/2 receptor antagonist, Sch527123, inhibits neutrophil recruitment, mucus production, and goblet cell hyperplasia in animal models of pulmonary inflammation. J Pharmacol Exp Ther 322: 486–493, 2007 [DOI] [PubMed] [Google Scholar]
  • 29.Cheng OZ, Palaniyar N. NET balancing: a problem in inflammatory lung diseases. Front Immunol 4: 1, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chignard M, Balloy V. Neutrophil recruitment and increased permeability during acute lung injury induced by lipopolysaccharide. Am J Physiol Lung Cell Mol Physiol 279: L1083–L1090, 2000 [DOI] [PubMed] [Google Scholar]
  • 31.Chollet-Martin S, Jourdain B, Gibert C, Elbim C, Chastre J, Gougerot-Pocidalo MA. Interactions between neutrophils and cytokines in blood and alveolar spaces during ARDS. Am J Respir Crit Care Med 154: 594–601, 1996 [DOI] [PubMed] [Google Scholar]
  • 32.Chollet-Martin S, Montravers P, Gibert C, Elbim C, Desmonts JM, Fagon JY, Gougerot-Pocidalo MA. High levels of interleukin-8 in the blood and alveolar spaces of patients with pneumonia and adult respiratory distress syndrome. Infect Immun 61: 4553–4559, 1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Christoffersson G, Vagesjo E, Vandooren J, Liden M, Massena S, Reinert RB, Brissova M, Powers AC, Opdenakker G, Phillipson M. VEGF-A recruits a proangiogenic MMP-9-delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue. Blood 120: 4653–4662, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chuntharapai A, Lee J, Hebert CA, Kim KJ. Monoclonal antibodies detect different distribution patterns of IL-8 receptor A and IL-8 receptor B on human peripheral blood leukocytes. J Immunol 153: 5682–5688, 1994 [PubMed] [Google Scholar]
  • 35.Clynes R, Maizes JS, Guinamard R, Ono M, Takai T, Ravetch JV. Modulation of immune complex-induced inflammation in vivo by the coordinate expression of activation and inhibitory Fc receptors. J Exp Med 189: 179–185, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Coldren CD, Nick JA, Poch KR, Woolum MD, Fouty BW, O'Brien JM, Gruber MP, Zamora MR, Svetkauskaite D, Richter DA, He Q, Park JS, Overdier KH, Abraham E, Geraci MW. Functional and genomic changes induced by alveolar transmigration in human neutrophils. Am J Physiol Lung Cell Mol Physiol 291: L1267–L1276, 2006 [DOI] [PubMed] [Google Scholar]
  • 37.Cordingley JL. Pores of Kohn. Thorax 27: 433–441, 1972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Costa C, Rufino R, Traves SL, Lapa e Silva JR, Barnes PJ, Donnelly LE. CXCR3 and CCR5 chemokines in induced sputum from patients with COPD. Chest 133: 26–33, 2008 [DOI] [PubMed] [Google Scholar]
  • 39.Crosby LM, Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol 298: L715–L731, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Cummings CJ, Martin TR, Frevert CW, Quan JM, Wong VA, Mongovin SM, Hagen TR, Steinberg KP, Goodman RB. Expression and function of the chemokine receptors CXCR1 and CXCR2 in sepsis. J Immunol 162: 2341–2346, 1999 [PubMed] [Google Scholar]
  • 41.Delclaux C, Rezaiguia-Delclaux S, Delacourt C, Brun-Buisson C, Lafuma C, Harf A. Alveolar neutrophils in endotoxin-induced and bacteria-induced acute lung injury in rats. Am J Physiol Lung Cell Mol Physiol 273: L104–L112, 1997 [DOI] [PubMed] [Google Scholar]
  • 42.Doerschuk CM, Allard MF, Martin BA, MacKenzie A, Autor AP, Hogg JC. Marginated pool of neutrophils in rabbit lungs. J Appl Physiol 63: 1806–1815, 1987 [DOI] [PubMed] [Google Scholar]
  • 43.Donnelly SC, MacGregor I, Zamani A, Gordon MW, Robertson CE, Steedman DJ, Little K, Haslett C. Plasma elastase levels and the development of the adult respiratory distress syndrome. Am J Respir Crit Care Med 151: 1428–1433, 1995 [DOI] [PubMed] [Google Scholar]
  • 44.Douda DN, Jackson R, Grasemann H, Palaniyar N. Innate immune collectin surfactant protein D simultaneously binds both neutrophil extracellular traps and carbohydrate ligands and promotes bacterial trapping. J Immunol 187: 1856–1865, 2011 [DOI] [PubMed] [Google Scholar]
  • 45.Dunican A, Grutkoski P, Leuenroth S, Ayala A, Simms HH. Neutrophils regulate their own apoptosis via preservation of CXC receptors. J Surg Res 90: 32–38, 2000 [DOI] [PubMed] [Google Scholar]
  • 46.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] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ferguson ND, Fan E, Camporota L, Antonelli M, Anzueto A, Beale R, Brochard L, Brower R, Esteban A, Gattinoni L, Rhodes A, Slutsky AS, Vincent JL, Rubenfeld GD, Thompson BT, Ranieri VM. The Berlin definition of ARDS: an expanded rationale, justification, and supplementary material. Intensive Care Med 38: 1573–1582, 2012 [DOI] [PubMed] [Google Scholar]
  • 48.Festic E, Ortiz-Diaz E, Lee A, Li G, Kor DJ, Adebola A, Akca O, Hoth J, Levitt JE, Carter R, Gajic O. Prehospital use of inhaled steroids and incidence of acute lung injury among patients at risk. J Crit Care 28: 985–991, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Folkesson HG, Matthay MA, Hebert CA, Broaddus VC. Acid aspiration-induced lung injury in rabbits is mediated by interleukin-8-dependent mechanisms. J Clin Invest 96: 107–116, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Fortunati E, Kazemier KM, Grutters JC, Koenderman L, Van den Bosch vJ Human neutrophils switch to an activated phenotype after homing to the lung irrespective of inflammatory disease. Clin Exp Immunol 155: 559–566, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fox ED, Heffernan DS, Cioffi WG, Reichner JS. Neutrophils from critically ill septic patients mediate profound loss of endothelial barrier integrity. Crit Care 17: R226, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fox SE, Lu W, Maheshwari A, Christensen RD, Calhoun DA. The effects and comparative differences of neutrophil specific chemokines on neutrophil chemotaxis of the neonate. Cytokine 29: 135–140, 2005 [DOI] [PubMed] [Google Scholar]
  • 53.Frevert CW, Huang S, Danaee H, Paulauskis JD, Kobzik L. Functional characterization of the rat chemokine KC and its importance in neutrophil recruitment in a rat model of pulmonary inflammation. J Immunol 154: 335–344, 1995 [PubMed] [Google Scholar]
  • 54.Fudala R, Krupa A, Matthay MA, Allen TC, Kurdowska AK. Anti-IL-8 autoantibody:IL-8 immune complexes suppress spontaneous apoptosis of neutrophils. Am J Physiol Lung Cell Mol Physiol 293: L364–L374, 2007 [DOI] [PubMed] [Google Scholar]
  • 55.Fudala R, Krupa A, Stankowska D, Allen TC, Kurdowska AK. Does activation of the FcgammaRIIa play a role in the pathogenesis of the acute lung injury/acute respiratory distress syndrome? Clin Sci (Lond) 118: 519–526, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Fujino N, Kubo H, Suzuki T, He M, Suzuki T, Yamada M, Takahashi T, Ota C, Yamaya M. Administration of a specific inhibitor of neutrophil elastase attenuates pulmonary fibrosis after acute lung injury in mice. Exp Lung Res 38: 28–36, 2012 [DOI] [PubMed] [Google Scholar]
  • 57.Gaggar A, Jackson PL, Noerager BD, O'Reilly PJ, McQuaid DB, Rowe SM, Clancy JP, Blalock JE. A novel proteolytic cascade generates an extracellular matrix-derived chemoattractant in chronic neutrophilic inflammation. J Immunol 180: 5662–5669, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Geiser T, Dewald B, Ehrengruber MU, Clark-Lewis I, Baggiolini M. The interleukin-8-related chemotactic cytokines GRO alpha, GRO beta, and GRO gamma activate human neutrophil and basophil leukocytes. J Biol Chem 268: 15419–15424, 1993 [PubMed] [Google Scholar]
  • 59.Gessner MA, Doran SF, Yu Z, Dunaway CW, Matalon S, Steele C. Chlorine gas exposure increases susceptibility to invasive lung fungal infection. Am J Physiol Lung Cell Mol Physiol 304: L765–L773, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ginzberg HH, Cherapanov V, Dong Q, Cantin A, McCulloch CA, Shannon PT, Downey GP. Neutrophil-mediated epithelial injury during transmigration: role of elastase. Am J Physiol Gastrointest Liver Physiol 281: G705–G717, 2001 [DOI] [PubMed] [Google Scholar]
  • 61.Goodman RB, Pugin J, Lee JS, Matthay MA. Cytokine-mediated inflammation in acute lung injury. Cytokine Growth Factor Rev 14: 523–535, 2003 [DOI] [PubMed] [Google Scholar]
  • 62.Goodman RB, Strieter RM, Frevert CW, Cummings CJ, Tekamp-Olson P, Kunkel SL, Walz A, Martin TR. Quantitative comparison of C-X-C chemokines produced by endotoxin-stimulated human alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 275: L87–L95, 1998 [DOI] [PubMed] [Google Scholar]
  • 63.Goodman RB, Strieter RM, Martin DP, Steinberg KP, Milberg JA, Maunder RJ, Kunkel SL, Walz A, Hudson LD, Martin TR. Inflammatory cytokines in patients with persistence of the acute respiratory distress syndrome. Am J Respir Crit Care Med 154: 602–611, 1996 [DOI] [PubMed] [Google Scholar]
  • 64.Gresnigt MS, Joosten LA, Verschueren I, Van Der Meer JW, Netea MG, Dinarello CA, Van De Veerdonk FL. Neutrophil-mediated inhibition of proinflammatory cytokine responses. J Immunol 189: 4806–4815, 2012 [DOI] [PubMed] [Google Scholar]
  • 65.Groeneveld AB, Raijmakers PG, Hack CE, Thijs LG. Interleukin 8-related neutrophil elastase and the severity of the adult respiratory distress syndrome. Cytokine 7: 746–752, 1995 [DOI] [PubMed] [Google Scholar]
  • 66.Grommes J, Alard JE, Drechsler M, Wantha S, Mörgelin M, Kuebler WM, Jacobs M, von Hundelshausen P, Markart P, Wygrecka M, Preissner KT, Hackeng TM, Koenen RR, Weber C, Soehnlein O. Disruption of platelet-derived chemokine heteromers prevents neutrophil extravasation in acute lung injury. Am J Respir Crit Care Med 185: 628–636, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Grommes J, Soehnlein O. Contribution of neutrophils to acute lung injury. Mol Med 17: 293–307, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gupta S, Feng L, Yoshimura T, Redick J, Fu SM, Rose CE., Jr Intra-alveolar macrophage-inflammatory peptide 2 induces rapid neutrophil localization in the lung. Am J Respir Cell Mol Biol 15: 656–663, 1996 [DOI] [PubMed] [Google Scholar]
  • 69.Harada A, Sekido N, Akahoshi T, Wada T, Mukaida N, Matsushima K. Essential involvement of interleukin-8 (IL-8) in acute inflammation. J Leukoc Biol 56: 559–564, 1994 [PubMed] [Google Scholar]
  • 70.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] [PubMed] [Google Scholar]
  • 71.Hauser CJ, Fekete Z, Goodman ER, Kleinstein E, Livingston DH, Deitch EA. CXCR2 stimulation primes CXCR1 [Ca2+]i responses to IL-8 in human neutrophils. Shock 12: 428–437, 1999 [DOI] [PubMed] [Google Scholar]
  • 72.Hayashida K, Parks WC, Park PW. Syndecan-1 shedding facilitates the resolution of neutrophilic inflammation by removing sequestered CXC chemokines. Blood 114: 3033–3043, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Herbold W, Maus R, Hahn I, Ding N, Srivastava M, Christman JW, Mack M, Reutershan J, Briles DE, Paton JC, Winter C, Welte T, Maus UA. Importance of CXC chemokine receptor 2 in alveolar neutrophil and exudate macrophage recruitment in response to pneumococcal lung infection. Infect Immun 78: 2620–2630, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hilgendorff A, Parai K, Ertsey R, Rey-Parra GJ, Thebaud B, Tamosiuniene R, Jain N, Navarro EF, Starcher BC, Nicolls MR, Rabinovitch M, Bland RD. Neonatal mice genetically modified to express the elastase inhibitor elafin are protected against the adverse effects of mechanical ventilation on lung growth. Am J Physiol Lung Cell Mol Physiol 303: L215–L227, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hill LL, Chen DL, Kozlowski J, Schuster DP. Neutrophils and neutrophil products do not mediate pulmonary hemodynamic effects of endotoxin on oleic acid-induced lung injury. Anesth Analg 98: 452–457, table, 2004 [DOI] [PubMed] [Google Scholar]
  • 76.Hirche TO, Atkinson JJ, Bahr S, Belaaouaj A. Deficiency in neutrophil elastase does not impair neutrophil recruitment to inflamed sites. Am J Respir Cell Mol Biol 30: 576–584, 2004 [DOI] [PubMed] [Google Scholar]
  • 77.Hogg JC. Neutrophil kinetics and lung injury. Physiol Rev 67: 1249–1295, 1987 [DOI] [PubMed] [Google Scholar]
  • 78.Holz O, Khalilieh S, Ludwig-Sengpiel A, Watz H, Stryszak P, Soni P, Tsai M, Sadeh J, Magnussen H. SCH527123, a novel CXCR2 antagonist, inhibits ozone-induced neutrophilia in healthy subjects. Eur Respir J 35: 564–570, 2010 [DOI] [PubMed] [Google Scholar]
  • 79.Hsieh CH, Frink M, Hsieh YC, Kan WH, Hsu JT, Schwacha MG, Choudhry MA, Chaudry IH. The role of MIP-1 alpha in the development of systemic inflammatory response and organ injury following trauma hemorrhage. J Immunol 181: 2806–2812, 2008 [DOI] [PubMed] [Google Scholar]
  • 80.Ichikawa A, Kuba K, Morita M, Chida S, Tezuka H, Hara H, Sasaki T, Ohteki T, Ranieri VM, Dos Santos CC, Kawaoka Y, Akira S, Luster AD, Lu B, Penninger JM, Uhlig S, Slutsky AS, Imai Y. CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin. Am J Respir Crit Care Med 187: 65–77, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Iwata K, Doi A, Ohji G, Oka H, Oba Y, Takimoto K, Igarashi W, Gremillion DH, Shimada T. Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): a systematic review and meta-analysis. Intern Med 49: 2423–2432, 2010 [DOI] [PubMed] [Google Scholar]
  • 82.Johnston B, Burns AR, Suematsu M, Issekutz TB, Woodman RC, Kubes P. Chronic inflammation upregulates chemokine receptors and induces neutrophil migration to monocyte chemoattractant protein-1. J Clin Invest 103: 1269–1276, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Johnston CJ, Finkelstein JN, Gelein R, Oberdorster G. Pulmonary cytokine and chemokine mRNA levels after inhalation of lipopolysaccharide in C57BL/6 mice. Toxicol Sci 46: 300–307, 1998 [DOI] [PubMed] [Google Scholar]
  • 84.Kamp VM, Pillay J, Lammers JW, Pickkers P, Ulfman LH, Koenderman L. Human suppressive neutrophils CD16bright/CD62Ldim exhibit decreased adhesion. J Leukoc Biol 92: 1011–1020, 2012 [DOI] [PubMed] [Google Scholar]
  • 85.Kantrow SP, Shen Z, Jagneaux T, Zhang P, Nelson S. Neutrophil-mediated lung permeability and host defense proteins. Am J Physiol Lung Cell Mol Physiol 297: L738–L745, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol 189: 2689–2695, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kasten KR, Muenzer JT, Caldwell CC. Neutrophils are significant producers of IL-10 during sepsis. Biochem Biophys Res Commun 393: 28–31, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kawabata K, Hagio T, Matsumoto S, Nakao S, Orita S, Aze Y, Ohno H. Delayed neutrophil elastase inhibition prevents subsequent progression of acute lung injury induced by endotoxin inhalation in hamsters. Am J Respir Crit Care Med 161: 2013–2018, 2000 [DOI] [PubMed] [Google Scholar]
  • 89.Kawano T, Mori S, Cybulsky M, Burger R, Ballin A, Cutz E, Bryan AC. Effect of granulocyte depletion in a ventilated surfactant-depleted lung. J Appl Physiol 62: 27–33, 1987 [DOI] [PubMed] [Google Scholar]
  • 90.Kaynar AM, Houghton AM, Lum EH, Pitt BR, Shapiro SD. Neutrophil elastase is needed for neutrophil emigration into lungs in ventilator-induced lung injury. Am J Respir Cell Mol Biol 39: 53–60, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 13: 159–175, 2013 [DOI] [PubMed] [Google Scholar]
  • 92.Konrad FM, Reutershan J. CXCR2 in acute lung injury. Mediators Inflamm 2012: 740987, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Krause PJ, Malech HL, Kristie J, Kosciol CM, Herson VC, Eisenfeld L, Pastuszak WT, Kraus A, Seligmann B. Polymorphonuclear leukocyte heterogeneity in neonates and adults. Blood 68: 200–204, 1986 [PubMed] [Google Scholar]
  • 94.Krishnadasan B, Farivar AS, Naidu BV, Woolley SM, Byrne K, Fraga CH, Mulligan MS. Beta-chemokine function in experimental lung ischemia-reperfusion injury. Ann Thorac Surg 77: 1056–1062, 2004 [DOI] [PubMed] [Google Scholar]
  • 95.Krupa A, Fudala R, Stankowska D, Loyd T, Allen TC, Matthay MA, Gryczynski Z, Gryczynski I, Mettikolla YV, Kurdowska AK. Anti-chemokine autoantibody:chemokine immune complexes activate endothelial cells via IgG receptors. Am J Respir Cell Mol Biol 41: 155–169, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Krupa A, Kato H, Matthay MA, Kurdowska AK. Proinflammatory activity of anti-IL-8 autoantibody:IL-8 complexes in alveolar edema fluid from patients with acute lung injury. Am J Physiol Lung Cell Mol Physiol 286: L1105–L1113, 2004 [DOI] [PubMed] [Google Scholar]
  • 97.Kubo H, Doyle NA, Graham L, Bhagwan SD, Quinlan WM, Doerschuk CM. L- and P-selectin and CD11/CD18 in intracapillary neutrophil sequestration in rabbit lungs. Am J Respir Crit Care Med 159: 267–274, 1999 [DOI] [PubMed] [Google Scholar]
  • 98.Kurdowska A, Miller EJ, Noble JM, Baughman RP, Matthay MA, Brelsford WG, Cohen AB. Anti-IL-8 autoantibodies in alveolar fluid from patients with the adult respiratory distress syndrome. J Immunol 157: 2699–2706, 1996 [PubMed] [Google Scholar]
  • 99.Kurdowska A, Noble JM, Grant IS, Robertson CR, Haslett C, Donnelly SC. Anti-interleukin-8 autoantibodies in patients at risk for acute respiratory distress syndrome. Crit Care Med 30: 2335–2337, 2002 [DOI] [PubMed] [Google Scholar]
  • 100.Kurdowska A, Noble JM, Steinberg KP, Ruzinski JT, Hudson LD, Martin TR. Anti-interleukin 8 autoantibody: interleukin 8 complexes in the acute respiratory distress syndrome. Relationship between the complexes and clinical disease activity. Am J Respir Crit Care Med 163: 463–468, 2001 [DOI] [PubMed] [Google Scholar]
  • 101.Kurdowska AK, Geiser TK, Alden SM, Dziadek BR, Noble JM, Nuckton TJ, Matthay MA. Activity of pulmonary edema fluid interleukin-8 bound to α2-macroglobulin in patients with acute lung injury. Am J Physiol Lung Cell Mol Physiol 282: L1092–L1098, 2002 [DOI] [PubMed] [Google Scholar]
  • 102.Lazaar AL, Sweeney LE, MacDonald AJ, Alexis NE, Chen C, Tal-Singer R. SB-656933, a novel CXCR2 selective antagonist, inhibits ex vivo neutrophil activation and ozone-induced airway inflammation in humans. Br J Clin Pharmacol 72: 282–293, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lee A, Whyte MK, Haslett C. Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators. J Leukoc Biol 54: 283–288, 1993 [PubMed] [Google Scholar]
  • 104.Leonard EJ, Yoshimura T. Neutrophil attractant/activation protein-1 (NAP-1 [interleukin-8]). Am J Respir Cell Mol Biol 2: 479–486, 1990 [DOI] [PubMed] [Google Scholar]
  • 105.Li B, Dong C, Wang G, Zheng H, Wang X, Bai C. Pulmonary epithelial CCR3 promotes LPS-induced lung inflammation by mediating release of IL-8. J Cell Physiol 226: 2398–2405, 2011 [DOI] [PubMed] [Google Scholar]
  • 106.Liang J, Jung Y, Tighe RM, Xie T, Liu N, Leonard M, Gunn MD, Jiang D, Noble PW. A macrophage subpopulation recruited by CC chemokine ligand-2 clears apoptotic cells in noninfectious lung injury. Am J Physiol Lung Cell Mol Physiol 302: L933–L940, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lin WC, Lin CF, Chen CL, Chen CW, Lin YS. Prediction of outcome in patients with acute respiratory distress syndrome by bronchoalveolar lavage inflammatory mediators. Exp Biol Med (Maywood) 235: 57–65, 2010 [DOI] [PubMed] [Google Scholar]
  • 108.Lomas JL, Chung CS, Grutkoski PS, LeBlanc BW, Lavigne L, Reichner J, Gregory SH, Doughty LA, Cioffi WG, Ayala A. Differential effects of macrophage inflammatory chemokine-2 and keratinocyte-derived chemokine on hemorrhage-induced neutrophil priming for lung inflammation: assessment by adoptive cells transfer in mice. Shock 19: 358–365, 2003 [DOI] [PubMed] [Google Scholar]
  • 109.Lomas-Neira JL, Ayala A. CXCL2 polymorphism in sepsis and acute respiratory distress syndrome: pathological significance lost in translation. Crit Care Med 35: 2439–2440, 2007 [DOI] [PubMed] [Google Scholar]
  • 110.Lomas-Neira JL, Chung CS, Wesche DE, Perl M, Ayala A. In vivo gene silencing (with siRNA) of pulmonary expression of MIP-2 versus KC results in divergent effects on hemorrhage-induced, neutrophil-mediated septic acute lung injury. J Leukoc Biol 77: 846–853, 2005 [DOI] [PubMed] [Google Scholar]
  • 111.Looney MR, Matthay MA. Animal models of transfusion-related acute lung injury. Crit Care Med 34: S132–S136, 2006 [DOI] [PubMed] [Google Scholar]
  • 112.Makita H, Nishimura M, Miyamoto K, Nakano T, Tanino Y, Hirokawa J, Nishihira J, Kawakami Y. Effect of anti-macrophage migration inhibitory factor antibody on lipopolysaccharide-induced pulmonary neutrophil accumulation. Am J Respir Crit Care Med 158: 573–579, 1998 [DOI] [PubMed] [Google Scholar]
  • 113.Marshall RP, Bellingan G, Webb S, Puddicombe A, Goldsack N, McAnulty RJ, Laurent GJ. Fibroproliferation occurs early in the acute respiratory distress syndrome and impacts on outcome. Am J Respir Crit Care Med 162: 1783–1788, 2000 [DOI] [PubMed] [Google Scholar]
  • 114.Martin TR, Pistorese BP, Chi EY, Goodman RB, Matthay MA. Effects of leukotriene B4 in the human lung. Recruitment of neutrophils into the alveolar spaces without a change in protein permeability. J Clin Invest 84: 1609–1619, 1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Matthay MA, Eschenbacher WL, Goetzl EJ. Elevated concentrations of leukotriene D4 in pulmonary edema fluid of patients with the adult respiratory distress syndrome. J Clin Immunol 4: 479–483, 1984 [DOI] [PubMed] [Google Scholar]
  • 116.Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest 122: 2731–2740, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Matute-Bello G, Liles WC, Radella F, 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] [PubMed] [Google Scholar]
  • 118.Maus UA, Waelsch K, Kuziel WA, Delbeck T, Mack M, Blackwell TS, Christman JW, Schlondorff D, Seeger W, Lohmeyer J. Monocytes are potent facilitators of alveolar neutrophil emigration during lung inflammation: role of the CCL2-CCR2 axis. J Immunol 170: 3273–3278, 2003 [DOI] [PubMed] [Google Scholar]
  • 119.McQuibban GA, Gong JH, Wong JP, Wallace JL, Clark-Lewis I, Overall CM. Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood 100: 1160–1167, 2002 [PubMed] [Google Scholar]
  • 120.Mehrad B, Strieter RM, Moore TA, Tsai WC, Lira SA, Standiford TJ. CXC chemokine receptor-2 ligands are necessary components of neutrophil-mediated host defense in invasive pulmonary aspergillosis. J Immunol 163: 6086–6094, 1999 [PubMed] [Google Scholar]
  • 121.Mei J, Liu Y, Dai N, Favara M, Greene T, Jeyaseelan S, Poncz M, Lee JS, Worthen GS. CXCL5 regulates chemokine scavenging and pulmonary host defense to bacterial infection. Immunity 33: 106–117, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Mei J, Liu Y, Dai N, Hoffmann C, Hudock KM, Zhang P, Guttentag SH, Kolls JK, Oliver PM, Bushman FD, Worthen GS. Cxcr2 and Cxcl5 regulate the IL-17/G-CSF axis and neutrophil homeostasis in mice. J Clin Invest 122: 974–986, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Mercer PF, Williams AE, Scotton CJ, Jose RJ, Sulikowski M, Moffatt JD, Murray LA, Chambers RC. Proteinase-activated receptor-1, CCL2 and CCL7 regulate acute neutrophilic lung inflammation. Am J Respir Cell Mol Biol 2013. August 23 [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Michalec L, Choudhury BK, Postlethwait E, Wild JS, Alam R, Lett-Brown M, Sur S. CCL7 and CXCL10 orchestrate oxidative stress-induced neutrophilic lung inflammation. J Immunol 168: 846–852, 2002 [DOI] [PubMed] [Google Scholar]
  • 125.Milks LC, Brontoli MJ, Cramer EB. Epithelial permeability and the transepithelial migration of human neutrophils. J Cell Biol 96: 1241–1247, 1983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Miller EJ, Cohen AB, Matthay MA. Increased interleukin-8 concentrations in the pulmonary edema fluid of patients with acute respiratory distress syndrome from sepsis. Crit Care Med 24: 1448–1454, 1996 [DOI] [PubMed] [Google Scholar]
  • 127.Miller EJ, Cohen AB, Nagao S, Griffith D, Maunder RJ, Martin TR, Weiner-Kronish JP, Sticherling M, Christophers E, Matthay MA. Elevated levels of NAP-1/interleukin-8 are present in the airspaces of patients with the adult respiratory distress syndrome and are associated with increased mortality. Am Rev Respir Dis 146: 427–432, 1992 [DOI] [PubMed] [Google Scholar]
  • 128.Mizgerd JP, Horwitz BH, Quillen HC, Scott ML, Doerschuk CM. Effects of CD18 deficiency on the emigration of murine neutrophils during pneumonia. J Immunol 163: 995–999, 1999 [PubMed] [Google Scholar]
  • 129.Moepps B, Nuesseler E, Braun M, Gierschik P. A homolog of the human chemokine receptor CXCR1 is expressed in the mouse. Mol Immunol 43: 897–914, 2006 [DOI] [PubMed] [Google Scholar]
  • 130.Monchi M, Bellenfant F, Cariou A, Joly LM, Thebert D, Laurent I, Dhainaut JF, Brunet F. Early predictive factors of survival in the acute respiratory distress syndrome. A multivariate analysis. Am J Respir Crit Care Med 158: 1076–1081, 1998 [DOI] [PubMed] [Google Scholar]
  • 131.Moreland JG, Fuhrman RM, Pruessner JA, Schwartz DA. CD11b and intercellular adhesion molecule-1 are involved in pulmonary neutrophil recruitment in lipopolysaccharide-induced airway disease. Am J Respir Cell Mol Biol 27: 474–480, 2002 [DOI] [PubMed] [Google Scholar]
  • 132.Mulligan MS, Vaporciyan AA, Warner RL, Jones ML, Foreman KE, Miyasaka M, Todd RF, III, Ward PA. Compartmentalized roles for leukocytic adhesion molecules in lung inflammatory injury. J Immunol 154: 1350–1363, 1995 [PubMed] [Google Scholar]
  • 133.Narasaraju T, Yang E, Samy RP, Ng HH, Poh WP, Liew AA, Phoon MC, van Rooijen N, Chow VT. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am J Pathol 179: 199–210, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Nash S, Stafford J, Madara JL. Effects of polymorphonuclear leukocyte transmigration on the barrier function of cultured intestinal epithelial monolayers. J Clin Invest 80: 1104–1113, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.O'Grady NP, Preas HL, Pugin J, Fiuza C, Tropea M, Reda D, Banks SM, Suffredini AF. Local inflammatory responses following bronchial endotoxin instillation in humans. Am J Respir Crit Care Med 163: 1591–1598, 2001 [DOI] [PubMed] [Google Scholar]
  • 136.O'Leary EC, Marder P, Zuckerman SH. Glucocorticoid effects in an endotoxin-induced rat pulmonary inflammation model: differential effects on neutrophil influx, integrin expression, and inflammatory mediators. Am J Respir Cell Mol Biol 15: 97–106, 1996 [DOI] [PubMed] [Google Scholar]
  • 137.Ognibene FP, Martin SE, Parker MM, Schlesinger T, Roach P, Burch C, Shelhamer JH, Parrillo JE. Adult respiratory distress syndrome in patients with severe neutropenia. N Engl J Med 315: 547–551, 1986 [DOI] [PubMed] [Google Scholar]
  • 138.Palmgren MS, deShazo RD, Carter RM, Zimny ML, Shah SV. Mechanisms of neutrophil damage to human alveolar extracellular matrix: the role of serine and metalloproteases. J Allergy Clin Immunol 89: 905–915, 1992 [DOI] [PubMed] [Google Scholar]
  • 139.Parker H, Dragunow M, Hampton MB, Kettle AJ, Winterbourn CC. Requirements for NADPH oxidase and myeloperoxidase in neutrophil extracellular trap formation differ depending on the stimulus. J Leukoc Biol 92: 841–849, 2012 [DOI] [PubMed] [Google Scholar]
  • 140.Parkos CA. Molecular events in neutrophil transepithelial migration. Bioessays 19: 865–873, 1997 [DOI] [PubMed] [Google Scholar]
  • 141.Parsons PE, Fowler AA, Hyers TM, Henson PM. Chemotactic activity in bronchoalveolar lavage fluid from patients with adult respiratory distress syndrome. Am Rev Respir Dis 132: 490–493, 1985 [DOI] [PubMed] [Google Scholar]
  • 142.Peao MN, Aguas AP, de Sa CM, Grande NR. Morphological evidence for migration of particle-laden macrophages through the interalveolar pores of Kohn in the murine lung. Acta Anat (Basel) 147: 227–232, 1993 [DOI] [PubMed] [Google Scholar]
  • 143.Perkowski S, Scherpereel A, Murciano JC, Arguiri E, Solomides CC, Albelda SM, Muzykantov V, Christofidou-Solomidou M. Dissociation between alveolar transmigration of neutrophils and lung injury in hyperoxia. Am J Physiol Lung Cell Mol Physiol 291: L1050–L1058, 2006 [DOI] [PubMed] [Google Scholar]
  • 144.Pignatti P, Moscato G, Casarini S, Delmastro M, Poppa M, Brunetti G, Pisati P, Balbi B. Downmodulation of CXCL8/IL-8 receptors on neutrophils after recruitment in the airways. J Allergy Clin Immunol 115: 88–94, 2005 [DOI] [PubMed] [Google Scholar]
  • 145.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] [PMC free article] [PubMed] [Google Scholar]
  • 146.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] [PubMed] [Google Scholar]
  • 147.Quinn DA, Moufarrej RK, Volokhov A, Hales CA. Interactions of lung stretch, hyperoxia, and MIP-2 production in ventilator-induced lung injury. J Appl Physiol 93: 517–525, 2002 [DOI] [PubMed] [Google Scholar]
  • 148.Reaves TA, Colgan SP, Selvaraj P, Pochet MM, Walsh S, Nusrat A, Liang TW, Madara JL, Parkos CA. Neutrophil transepithelial migration: regulation at the apical epithelial surface by Fc-mediated events. Am J Physiol Gastrointest Liver Physiol 280: G746–G754, 2001 [DOI] [PubMed] [Google Scholar]
  • 149.Reutershan J, Harry B, Chang D, Bagby GJ, Ley K. DARC on RBC limits lung injury by balancing compartmental distribution of CXC chemokines. Eur J Immunol 39: 1597–1607, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Reutershan J, Morris MA, Burcin TL, Smith DF, Chang D, Saprito MS, Ley K. Critical role of endothelial CXCR2 in LPS-induced neutrophil migration into the lung. J Clin Invest 116: 695–702, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ, Hudson LD. Incidence and outcomes of acute lung injury. N Engl J Med 353: 1685–1693, 2005 [DOI] [PubMed] [Google Scholar]
  • 152.Schmal H, Shanley TP, Jones ML, Friedl HP, Ward PA. Role for macrophage inflammatory protein-2 in lipopolysaccharide-induced lung injury in rats. J Immunol 156: 1963–1972, 1996 [PubMed] [Google Scholar]
  • 153.Sekido N, Mukaida N, Harada A, Nakanishi I, Watanabe Y, Matsushima K. Prevention of lung reperfusion injury in rabbits by a monoclonal antibody against interleukin-8. Nature 365: 654–657, 1993 [DOI] [PubMed] [Google Scholar]
  • 154.Shanley TP, Davidson BA, Nader ND, Bless N, Vasi N, Ward PA, Johnson KJ, Knight PR. Role of macrophage inflammatory protein-2 in aspiration-induced lung injury. Crit Care Med 28: 2437–2444, 2000 [DOI] [PubMed] [Google Scholar]
  • 155.Shanley TP, Schmal H, Friedl HP, Jones ML, Ward PA. Role of macrophage inflammatory protein-1 alpha (MIP-1 alpha) in acute lung injury in rats. J Immunol 154: 4793–4802, 1995 [PubMed] [Google Scholar]
  • 156.Simon RH, DeHart PD, Todd RF., III Neutrophil-induced injury of rat pulmonary alveolar epithelial cells. J Clin Invest 78: 1375–1386, 1986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sirianni FE, Chu FS, Walker DC. Human alveolar wall fibroblasts directly link epithelial type 2 cells to capillary endothelium. Am J Respir Crit Care Med 168: 1532–1537, 2003 [DOI] [PubMed] [Google Scholar]
  • 158.Snelgrove RJ. Targeting of a common receptor shared by CXCL8 and N-Ac-PGP as a therapeutic strategy to alleviate chronic neutrophilic lung diseases. Eur J Pharmacol 667: 1–5, 2011 [DOI] [PubMed] [Google Scholar]
  • 159.Soehnlein O, Oehmcke S, Ma X, Rothfuchs AG, Frithiof R, van RN, Morgelin M, Herwald H, Lindbom L. Neutrophil degranulation mediates severe lung damage triggered by streptococcal M1 protein. Eur Respir J 32: 405–412, 2008 [DOI] [PubMed] [Google Scholar]
  • 160.Song J, Wu C, Zhang X, Sorokin LM. In vivo processing of CXCL5 (LIX) by matrix metalloproteinase (MMP)-2 and MMP-9 promotes early neutrophil recruitment in IL-1beta-induced peritonitis. J Immunol 190: 401–410, 2013 [DOI] [PubMed] [Google Scholar]
  • 161.Standiford TJ, Kunkel SL, Lukacs NW, Greenberger MJ, Danforth JM, Kunkel RG, Strieter RM. Macrophage inflammatory protein-1 alpha mediates lung leukocyte recruitment, lung capillary leak, and early mortality in murine endotoxemia. J Immunol 155: 1515–1524, 1995 [PubMed] [Google Scholar]
  • 162.Steinberg KP, Milberg JA, Martin TR, Maunder RJ, Cockrill BA, Hudson LD. Evolution of bronchoalveolar cell populations in the adult respiratory distress syndrome. Am J Respir Crit Care Med 150: 113–122, 1994 [DOI] [PubMed] [Google Scholar]
  • 163.Struyf S, Gouwy M, Dillen C, Proost P, Opdenakker G, Van DJ. Chemokines synergize in the recruitment of circulating neutrophils into inflamed tissue. Eur J Immunol 35: 1583–1591, 2005 [DOI] [PubMed] [Google Scholar]
  • 164.Sue RD, Belperio JA, Burdick MD, Murray LA, Xue YY, Dy MC, Kwon JJ, Keane MP, Strieter RM. CXCR2 is critical to hyperoxia-induced lung injury. J Immunol 172: 3860–3868, 2004 [DOI] [PubMed] [Google Scholar]
  • 165.Swee M, Wilson CL, Wang Y, McGuire JK, Parks WC. Matrix metalloproteinase-7 (matrilysin) controls neutrophil egress by generating chemokine gradients. J Leukoc Biol 83: 1404–1412, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Sylvester I, Yoshimura T, Sticherling M, Schroder JM, Ceska M, Peichl P, Leonard EJ. Neutrophil attractant protein-1-immunoglobulin G immune complexes and free anti-NAP-1 antibody in normal human serum. J Clin Invest 90: 471–481, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.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] [PMC free article] [PubMed] [Google Scholar]
  • 168.Thomas GM, Carbo C, Curtis BR, Martinod K, Mazo IB, Schatzberg D, Cifuni SM, Fuchs TA, von Andrian UH, Hartwig JH, Aster RH, Wagner DD. Extracellular DNA traps are associated with the pathogenesis of TRALI in humans and mice. Blood 119: 6335–6343, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Thommasen HV, Russell JA, Boyko WJ, Hogg JC. Transient leucopenia associated with adult respiratory distress syndrome. Lancet 1: 809–812, 1984 [DOI] [PubMed] [Google Scholar]
  • 170.Tkalcevic J, Novelli M, Phylactides M, Iredale JP, Segal AW, Roes J. Impaired immunity and enhanced resistance to endotoxin in the absence of neutrophil elastase and cathepsin G. Immunity 12: 201–210, 2000 [DOI] [PubMed] [Google Scholar]
  • 171.Tosi MF, Stark JM, Hamedani A, Smith CW, Gruenert DC, Huang YT. Intercellular adhesion molecule-1 (ICAM-1)-dependent and ICAM-1-independent adhesive interactions between polymorphonuclear leukocytes and human airway epithelial cells infected with parainfluenza virus type 2. J Immunol 149: 3345–3349, 1992 [PubMed] [Google Scholar]
  • 172.Tosi MF, Stark JM, Smith CW, Hamedani A, Gruenert DC, Infeld MD. Induction of ICAM-1 expression on human airway epithelial cells by inflammatory cytokines: effects on neutrophil-epithelial cell adhesion. Am J Respir Cell Mol Biol 7: 214–221, 1992 [DOI] [PubMed] [Google Scholar]
  • 173.Tsuda Y, Takahashi H, Kobayashi M, Hanafusa T, Herndon DN, Suzuki F. Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus. Immunity 21: 215–226, 2004 [DOI] [PubMed] [Google Scholar]
  • 174.Urban CF, Reichard U, Brinkmann V, Zychlinsky A. Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol 8: 668–676, 2006 [DOI] [PubMed] [Google Scholar]
  • 175.Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing, whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood 96: 2673–2681, 2000 [PubMed] [Google Scholar]
  • 176.Villard J, Dayer-Pastore F, Hamacher J, Aubert JD, Schlegel-Haueter S, Nicod LP. GRO alpha and interleukin-8 in Pneumocystis carinii or bacterial pneumonia and adult respiratory distress syndrome. Am J Respir Crit Care Med 152: 1549–1554, 1995 [DOI] [PubMed] [Google Scholar]
  • 177.Walker DC, Behzad AR, Chu F. Neutrophil migration through preexisting holes in the basal laminae of alveolar capillaries and epithelium during streptococcal pneumonia. Microvasc Res 50: 397–416, 1995 [DOI] [PubMed] [Google Scholar]
  • 178.Wang XQ, Zhou X, Zhou Y, Rong L, Gao L, Xu W. Low-dose dexamethasone alleviates lipopolysaccharide-induced acute lung injury in rats and upregulates pulmonary glucocorticoid receptors. Respirology 13: 772–780, 2008 [DOI] [PubMed] [Google Scholar]
  • 179.Weathington NM, van Houwelingen AH, Noerager BD, Jackson PL, Kraneveld AD, Galin FS, Folkerts G, Nijkamp FP, Blalock JE. A novel peptide CXCR ligand derived from extracellular matrix degradation during airway inflammation. Nat Med 12: 317–323, 2006 [DOI] [PubMed] [Google Scholar]
  • 180.Wiedermann FJ, Mayr AJ, Kaneider NC, Fuchs D, Mutz NJ, Schobersberger W. Alveolar granulocyte colony-stimulating factor and alpha-chemokines in relation to serum levels, pulmonary neutrophilia, and severity of lung injury in ARDS. Chest 125: 212–219, 2004 [DOI] [PubMed] [Google Scholar]
  • 181.Wilhelmsen K, Mesa KR, Prakash A, Xu F, Hellman J. Activation of endothelial TLR2 by bacterial lipoprotein upregulates proteins specific for the neutrophil response. Innate Immun 18: 602–616, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.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] [PMC free article] [PubMed] [Google Scholar]
  • 183.Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, Taylor FB, Esmon NL, Lupu F, Esmon CT. Extracellular histones are major mediators of death in sepsis. Nat Med 15: 1318–1321, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Xu X, Jackson PL, Tanner S, Hardison MT, Abdul RM, Blalock JE, Gaggar A. A self-propagating matrix metalloprotease-9 (MMP-9) dependent cycle of chronic neutrophilic inflammation. PLoS One 6: e15781, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Yamada M, Kubo H, Kobayashi S, Ishizawa K, He M, Suzuki T, Fujino N, Kunishima H, Hatta M, Nishimaki K, Aoyagi T, Tokuda K, Kitagawa M, Yano H, Tamamura H, Fujii N, Kaku M. The increase in surface CXCR4 expression on lung extravascular neutrophils and its effects on neutrophils during endotoxin-induced lung injury. Cell Mol Immunol 8: 305–314, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Yamamoto T, Kajikawa O, Martin TR, Sharar SR, Harlan JM, Winn RK. The role of leukocyte emigration and IL-8 on the development of lipopolysaccharide-induced lung injury in rabbits. J Immunol 161: 5704–5709, 1998 [PubMed] [Google Scholar]
  • 187.Yamasawa H, Ishii Y, Kitamura S. Cytokine-induced neutrophil chemoattractant in a rat model of lipopolysaccharide-induced acute lung injury. Inflammation 23: 263–274, 1999 [DOI] [PubMed] [Google Scholar]
  • 188.Yang D, Tong L, Wang D, Wang Y, Wang X, Bai C. Roles of CC chemokine receptors (CCRs) on lipopolysaccharide-induced acute lung injury. Respir Physiol Neurobiol 170: 253–259, 2010 [DOI] [PubMed] [Google Scholar]
  • 189.Yokoi K, Mukaida N, Harada A, Watanabe Y, Matsushima K. Prevention of endotoxemia-induced acute respiratory distress syndrome-like lung injury in rabbits by a monoclonal antibody to IL-8. Lab Invest 76: 375–384, 1997 [PubMed] [Google Scholar]
  • 190.Zarbock A, Allegretti M, Ley K. Therapeutic inhibition of CXCR2 by Reparixin attenuates acute lung injury in mice. Br J Pharmacol 155: 357–364, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Zemans RL, Briones N, Campbell M, McClendon J, Young SK, Suzuki T, Yang IV, De LS, Reynolds SD, Mason RJ, Kahn M, Henson PM, Colgan SP, Downey GP. Neutrophil transmigration triggers repair of the lung epithelium via beta-catenin signaling. Proc Natl Acad Sci USA 108: 15990–15995, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Zemans RL, Colgan SP, Downey GP. Transepithelial migration of neutrophils: mechanisms and implications for acute lung injury. Am J Respir Cell Mol Biol 40: 519–535, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Zemans RL, McClendon J, Aschner Y, Briones N, Young SK, Lau LF, Kahn M, Downey GP. Role of beta-catenin-regulated CCN matricellular proteins in epithelial repair after inflammatory lung injury. Am J Physiol Lung Cell Mol Physiol 304: L415–L427, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zisman DA, Kunkel SL, Strieter RM, Tsai WC, Bucknell K, Wilkowski J, Standiford TJ. MCP-1 protects mice in lethal endotoxemia. J Clin Invest 99: 2832–2836, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

RESOURCES