SHOCK, Vol. 41, No. 6, pp. 491Y498, 2014

Review Article ROLE OF NEUTROPHIL EXTRACELLULAR TRAPS FOLLOWING INJURY Fu-Chao Liu,*† Yueh-Hsun Chuang,‡ Yung-Fong Tsai,*† and Huang-Ping Yu*† *Department of Anesthesiology, Chang Gung Memorial Hospital; and † College of Medicine, Chang Gung University, Taoyuan; and ‡ Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan Received 28 Nov 2013; first review completed 17 Dec 2013; accepted in final form 22 Jan 2014 ABSTRACT—Neutrophil extracellular traps (NETs), which consist of neutrophil DNA and cytoplasmic proteins, have been shown to be involved in various infectious, inflammatory, and autoimmune diseases. Neutrophil extracellular traps are abundant at the site of infection and acute inflammation. Neutrophil extracellular trap formation can occur through various intracellular signaling pathways, including peptidylarginine deiminase 4, Raf-MEK-ERK, nitric oxide, Toll-like receptor 4, high mobility group box 1, pentraxin 3, and mammalian targets of rapamycin. A growing body of evidence indicates that NETs may play an important role in injury, and decreases in NETs could reduce tissue injury. Neutrophil extracellular traps are believed to modulate the inflammatory and immune responses of individuals after injury. In this review, the role of NETs in injury, including traumatic injury, ischemia-reperfusionYinduced injury, and sepsis, as well as the potential markers and therapeutic targets of NET-related injury will be discussed. KEYWORDS—Neutrophil extracellular trap, injury

INTRODUCTION

severe inflammatory response and increase the risk of development of sepsis (22, 23). Neutrophils play a pivotal role in this inflammatory component after injury. The processes of neutrophilinduced tissue injury include proinflammatory mediator release, oxygen free radical generation, proteases degranulation, and endothelial dysfunction (2, 24). Activated neutrophils can release a number of cytokines and chemokines (e.g., interleukin 8 [IL-8], macrophage inflammatory protein 1!, etc.) (25, 26), and proinflammatory mediators (e.g., IL-6, tumor necrosis factor ! [TNF-!], IL-1"), and their secretion per se is regulated by immunoregulatory cytokines (27Y29). These mediators and factors may participate in recruiting more neutrophils or other leukocytes to the site of infection or inflammation. Neutrophil activation can be induced by host mediators (e.g., IL-8, platelet-activating factor, and TNF-!) and pathogens (e.g., formylated peptide and lipopolysaccharide [LPS]) (28, 30Y32). Neutrophils can also fight against microorganisms by directly phagocytosing microbes or releasing cytotoxic molecules via degranulation (33, 34). Phagocytosis is the process by which neutrophils directly engulf and digest potential pathogens, as well as cellular debris. Internalized pathogens are contained in phagosomes, where antimicrobial proteins from cellular granules and reactive oxygen species (ROS) produced by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase work together to create a toxic environment for invading pathogens (35, 36). Degranulation is the release of toxic ROS and antimicrobial granular proteins into the extracellular space. These granules contain antimicrobial proteins that fight infections in organs, including proteases, collagenases, lipoxygenases, phospholipases, and myeloperoxidase (MPO) (33, 37). Aside from phagocytosis and degranulation, neutrophils can also produce NETs during inflammation and infection (14, 17, 19, 38).

Neutrophils are the principal cells involved in host defense against microbial pathogen infections in the innate immune system (1, 2). Neutrophils also play a key role in the inflammatory response to injury (2, 3). Neutrophils are activated following injury, which is associated with an increased neutrophil accumulation in the damaged tissue or organ (4Y7). Activated neutrophils infiltrate injured tissue following an injury-induced increase in the expression of adhesion molecules on endothelial cells and elevated local chemokine/cytokine levels (6Y8). Severe injuries induce immune suppression, predisposing victims to postinjury complications (9Y12). Neutrophils enter tissues and organs following an injury or during an infection and form neutrophil extracellular traps (NETs) (13Y16). Neutrophil extracellular traps consist of neutrophil DNA, granular proteins, and several cytoplasmic proteins (17Y19). Recent reports indicate that NETs may contribute to tissue damage and organ dysfunction (13, 15, 20, 21). This review provides a brief overview of the role of NETs in injury and discusses the potential mechanisms by which NETs are involved in organ function and immunity modulation following an injury. In addition, the proposed markers and therapeutic targets of NET-related injury will be discussed. ROLE OF NEUTROPHILS IN TISSUE INJURY Studies have shown that tissue or organ injury, including trauma and ischemia-reperfusion (IR)Yinduced injury, can induce a Address reprint requests to Huang-Ping Yu, MD, PhD, Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fu-Shin St, Kwei-Shan, Tao-Yuan, Taiwan 333. E-mail: [email protected]. This work was partially supported by grants from the National Science Council (NSC102-2314-B-182A-051-MY3) and Chang Gung Memorial Hospital (CMRPG3B1052) to H.-P.Y. Support was also provided by the National Science Council (NSC101-2314-B-182-082) and Chang Gung Memorial Hospital (CMRPG3B1622) to F.-C.L. DOI: 10.1097/SHK.0000000000000146 Copyright Ó 2014 by the Shock Society

Extracellular traps

Extracellular DNA traps have been investigated in the context of neutrophil antimicrobial mechanisms, but not exclusively 491

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

492

SHOCK VOL. 41, NO. 6

focusing on neutrophils, as other cells also release extracellular traps composed of DNA and antimicrobial proteins. Extracellular DNA traps can be generated by macrophages, eosinophils, and mast cells (14, 39, 40). NET formation (NETosis)

Neutrophil extracellular traps contain granules and antimicrobial proteins, including histones, neutrophil elastase, MPO, pentraxin (PTX), lactoferrin, cathepsin G, and bactericidal permeabilityincreasing protein (17Y19). Neutrophil extracellular trap formation, referred to as NETosis, was first described by Brinkmann and colleagues as a cell death pathway distinct from apoptosis and necrosis (18, 19). NETosis leads to the dissolution of the nuclear envelop and cytoplasmic granules, allowing chromatin to mix with granular antimicrobial proteins via the NADPH oxidase and RAfMEK-ERK pathway (41). Subsequently, this mixture of granules, DNA, and histones is actively expelled from neutrophils into the extracellular environment, which limits the spreading of microbial pathogens. NETosis can be a protective process that sequesters microbes and prevents the spread of infection, but it can also be a pathological process that causes inflammation and serious tissue injury (42, 43). Although NETs play important roles in host defense by trapping bacteria or other pathogens, extensive formation of NETs with increased amounts of extracellular DNA may contribute to the perpetuation of inflammation and tissue damage (42, 44). Suicidal NETosis and vital NETosis

Recent study has reported that there are two different mechanisms by which NETs are formed, including a suicidal (lytic) NETosis and a vital (live cell) NETosis (45). Suicidal NETosis results from a developing membrane rupture and the loss of activating neutrophil functions, such as leukocyte phagocytosis, recruitment, and chemotaxis. The pathway of suicidal NETosis requires hours in the timing of NET release in response to phorbol 12 myristate 13 acetate (PMA) stimulation (46). In addition, MPO and neutrophil elastase mediate the chromatin decondensation leading to DNA and granule proteins mixing within the NET vacuole, then extruded out of a perforation in the plasma membrane (45). In vital NETosis, NET release is rapidly induced by LPS, and the pathogen-associated recognition occurs through host pattern recognition receptors. This rapid NETosis did not involve cell lysis and was mediated by platelets and Tolllike receptor 4 (TLR4) involved in the activation of polymorphonuclear neutrophils (47).

LIU ET

excess of NETosis can lead to tissue damage (17, 44, 45, 53). Although NETs provide an important biological advantage for the host to protect against certain microbial infections, the generation of NETs may be a double-edged sword. For example, although NETs may promote the destruction of pathogens, the same pathways may also cause injury through an overexpression of NETosis (20, 53). Neutrophil extracellular trap constituents can damage epithelial and endothelial cells, which can exacerbate inflammation-induced organ injury (17, 44, 54, 55). NETs and organ injury

It is well known that NET formation results in an extracellular release of proteases and other neutrophil constituents that can exacerbate injury (44). Several studies suggested that the magnitude of tissue damage and organ dysfunction are associated with the degree of NET formation (15Y17, 21) (Table 1). In human studies, NETs are present in the plasma of patients with acute lung injury and appear in the lung and plasma with transfusion-related acute lung injury (TRALI) (21). Serum NET levels were significantly increased immediately or 4 weeks after transplantation and were associated with an increased risk of transplantation-associated thrombotic microangiopathy (TA-TMA) (56). Neutrophil extracellular traps are also expressed in pulmonary capillaries and hepatic sinusoids during endotoxic shock (47). In animal studies, NETs are found in the alveoli of mice experiencing antibody-mediated TRALI. Deoxyribonuclease 1 (DNase 1) inhalation prevented the accumulation of NETs in alveoli and improved arterial oxygen saturation after TRALI (15). A recent study has shown that NETs are detected in the reperfused hind limb skeletal muscle and thrombosed vessels of wild-type mice (13). When DNase was used as a therapeutic agent to degrade the NETs, the results showed that there was a marked decrease in the NET levels detected in the muscle fibers, perivascular space, and microvascular thrombi of the ischemicreperfused hind limb following DNase treatment (13). In a mouse model of LPS-induced acute lung injury, NET formation appeared in the lung tissue, as well as bronchoalveolar lavage fluid. These findings reveal the important role of the protein components of NETs, particularly histones, which may lead to host cell cytotoxicity and be involved in lung tissue destruction. TABLE 1. Association of NETs in organ injury Species

Target organ

References

TRALI

Human, mice

Lung

Thomas et al. (21)

TRALI

Mice

Lung

Caudrillier et al. (15)

IR

Mice

Muscle

Oklu et al. (13)

LPS

Mice

lung

Saffarzadeh et al. (57)

Influenza A virus

Mice

Lung

Narasaraju et al. (16)

MCAO

Mice

Brain

Allen et al. (58)

MCAO

Mice

Brain

Enzmann et al. (60)

Damage

Activators of NETosis

Neutrophil extracellular traps are formed in response to biological and chemical stimuli, including IL-8, LPS, PMA, interferon +, TNF-!, activated platelets, and interaction with endothelial cells, bacteria, fungi, and complement-mediated opsonization (17Y19, 48), as well as through enhanced ROS generation by NADPH oxidase (49). Neutrophil extracellular traps and NET-like structures exist in many organisms, including humans (14, 50), mice, and other animals (39, 51, 52). ROLE OF NETS IN INJURY Neutrophil function and NETs are critical components involved in human immune defense. Although NETs are important, an

AL.

Sepsis

Human

Blood

Clark et al. (47)

TA-TMA

Human

Vessels

Arai et al. (56)

Mice

Vessels

Brill et al. (51)

Inferior vena cava stenosis

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

SHOCK JUNE 2014

NEUTROPHIL EXTRACELLULAR TRAPS FOLLOWING INJURY

Histones and MPO are responsible for NET-mediated cytotoxicity (57). When coincubated with infected alveolar epithelial cells in vitro, neutrophils from infected lungs strongly induce NET generation and augment endothelial damage (16). Another study has shown that transmigrated neutrophils release NETs following a transient middle cerebral artery occlusion (MCAO) in mice. Furthermore, a blockade of histone-DNA complexes attenuated transmigrated neutrophil-induced neuronal death, whereas inhibition of neutrophil proteases released and decondensed DNA in the brain (58). Previous study also showed that the IR-related damage is decreased in TLR4 mutant mice (C3H/HeJ) compared with wild-type mice following MCAO injuries (59). In contrast, Enzmann and colleagues (60) reported that absence of polymorphonuclear leukocyte infiltration and NET formation in the infarcted brain tissues after transient MCAO. Recent studies suggest that NETs are related to thrombosis and can damage the endothelium of blood vessels (51, 61, 62). Neutrophil extracellular trapYinduced thrombosis may play an important role in the pathogenesis of sepsis with reduced blood flow. Neutrophil extracellular trap production requires plateletneutrophil interactions and can be inhibited by platelet depletion or disruption of integrin-mediated platelet-neutrophil binding. During sepsis, NET release increases bacterial trapping by 4-fold.

493

Blocking NET formation reduces the capture of circulating bacteria during sepsis, resulting in an increased dissemination to distant organs (51, 61, 62). Neutrophil extracellular trap constituents can activate platelets and promote an excessive coagulopathy and thrombosis, resulting in endothelial cell injury and organ damage (63). NETs-related mediators and signaling pathways in injury

Neutrophil extracellular traps play an important role in both host defense and organ injury. Neutrophil extracellular trap formation is primarily dependent on histone levels, activation of NADPH oxidase and MPO, interactions between platelets and neutrophils, expression of NET component proteins, and neutrophil autophagy (15, 17, 18). The signaling molecules in NET formation include peptidylarginine deiminase 4 (PAD4) (64), Raf-MEK-ERK (41), nitric oxide (NO) (65), TLR4 (13), high mobility group box 1 (HMGB1) (66), and mammalian target of rapamycin (mTOR) (67) (Fig. 1). Peptidylarginine deiminase 4

Chromatin decondensation, which occurs in the nucleus, is a critical step in NET formation. Peptidylarginine deiminase 4 is a nuclear enzyme that converts specific arginine residues to citrulline

FIG. 1. Neutrophil extracellular trapsYrelated mediators and signaling pathways in injury. The signaling mechanisms of control NET formation may be mediated via Raf-MEK-ERK and NO pathway regulation NADPH oxidase and ROS expression. Toll-like receptor 4Yactivated platelets induce platelet-neutrophil interactions, resulting in NET formation. High mobility group box 1 can induce NETs through a TLR4-dependent pathway. Peptidylarginine deiminase 4Ymediated histone citrullination induces chromatin decondensation in NET formation. The mTOR kinase plays an essential role in NET release by regulation of autophagy. IFN indicates interferon; PI3K, phosphoinositide 3-kinase.

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

494

SHOCK VOL. 41, NO. 6

LIU ET

AL.

on histone tails (histone H3). The release of NETs depends on PAD4 activity (68, 69). Peptidylarginine deiminase 4 is activated by inflammatory stimuli, which is dependent on cell surface and cytoskeleton signaling (70). Neutrophils cannot release NETs in PAD4-mutant mice (64). Previous studies have shown that PAD4-mediated chromatin decondensation in neutrophils is critical in the pathogenesis of venous thrombosis (71).

mTOR-dependent NET is sensitive to inhibition of respiratory burst or blockade of cytoskeletal dynamics. The mTOR pathway is implicated in coordinating intracellular signaling of neutrophil activation related to NETosis (79, 80). Recently, mTOR was shown to regulate LPS-induced NET release by posttranscriptional control of hypoxia-inducible factor 1 (80).

Raf-MEK-ERK pathway

Other factors that influence NET formation

The Raf-MEK-ERK signaling pathway is a common pathway through which different stimuli can induce NETosis. Previous studies have shown that the Raf/MEK/ERK signaling pathway is critical for PMA-induced NET formation (71, 72). Interruption of MEK signaling reduces NET formation in platelet activation. Because ROS production is essential to NET formation, and the Raf-MEK-ERK pathway is the upstream of NADPH oxidase, it is involved in NET formation. In a molecular biology experiment, it was demonstrated that the Raf-MEK-ERK pathway appears to be involved in NET formation through the activation of NADPH oxidase and upregulation of antiapoptotic proteins (41).

In addition to the factors described above, several proinflammatory mediators and complements, such as IL-8, TNF-!, interferon, and complement 5, can stimulate the formation of NETs via NADPH oxidase signaling (55).

Nitric oxide

Nitric oxideYmediated NETs are important for nuclear and mitochondrial DNA, as well as for proteolytic enzymes. Previous studies have shown that augmenting NO with enzymatic free radical generation results in the release of NETs. These NETs are made up of mitochondrial and nuclear DNA and release proinflammatory cytokines (65, 73). Toll-like receptor 4

Previous study showed that LPS could stimulate HMGB1 expression and contribute to NET formation. High mobility group box 1 can induce NET formation both in vitro and in vivo through a TLR4-dependent mechanism (13, 74). In thrombosis, NETs are reported to have potential roles in clot formation via the TLR4 pathway (71, 75). Deletion of TLR4 from platelets dramatically reduces the formation of NETs (71, 75). Recently, it was shown that platelet TLR4 expression was essential for NET formation during endotoxic shock (47). In sepsis, TLR4-activated platelets induce platelet-neutrophil interactions, resulting in NET formation in blood vessels, especially in pulmonary capillaries and liver sinusoids (47). High mobility group box 1

High mobility group box 1 may contribute to neutrophilmediated tissue damage and organ dysfunction during acute inflammatory processes. High mobility group box 1 also plays a beneficial role in microbial eradication through its proinflammatory action and modulation of neutrophil chemotaxis (76, 77). Previous studies have shown that HMGB1 can potentiate NET formation, suggesting a novel mechanism by which HMGB1 may enhance host defense against bacterial infection and contribute to inflammatory processes (74, 78). Mammalian target of rapamycin

Mammalian target of rapamycin kinase may play an essential role in NET release by regulating autophagy (79, 80). Previous studies have shown that pharmacological inhibition of the mTOR pathway may enhance the rate of NET release. The release of

NET-ASSOCIATED PREDICTIVE MARKERS IN INJURY Cell-free DNA/NETs

Previous studies have shown that circulating cell-free DNA (cf-DNA)/NET levels can be potentially used for calculating injury severity following trauma with sepsis (81Y83). Clinical studies have also shown that cf-DNA/NET levels are important for predicting posttraumatic complications in the intensive care unit setting (83). Serum levels of cf-DNA/NETs are increased in trauma patients who subsequently develop sepsis (81). Cell-free DNA/NET levels in synovial fluid are also increased in patients with septic arthritis (82). Cell-free DNA/NET levels appear to be a valuable marker for the diagnosis of septic arthritis or periprosthetic infections. In an incremental treadmill test study, cf-DNA expression was shown to rapidly increase compared with other skeletal muscle damage markers, such as creatine kinase, uric acid, and C-reactive protein, suggesting that cf-DNA may be an important molecular marker in exercise physiology (84). In another clinical study, serum NET levels were significantly increased after TA-TMA (56). Increased NETs are an important risk factor for TA-TMA, suggesting that NET levels are a useful biomarker for TA-TMA. DNase

DNase is naturally present in human blood (85) and produced as a defense mechanism associated with NETs. The expression of DNase is significantly increased in the early phase of sepsis after major trauma (81). DNase degrades NETs in a concentrationdependent manner, and levels of DNase have been suggested to be a potential biomarker of NET formation following injuries (81). Pentraxin 3

Pentraxin 3 is a key protein component of NETs (85, 86) and mainly acts as a soluble pattern recognition receptor in the innate immune response (87, 88). Pentraxin 3 forms a complex with some of the other components of NETs (89) and appears to be an important molecule involved in enhancing the actions of other NET component proteins (86, 89). The association of circulating PTX3 with NET component proteins in sepsis suggests that it may be a potential diagnostic target (89). The PTX3 protein may also contribute to cell-mediated immune defense in patients with ulcerative colitis and crypt abscess lesions (90).

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

SHOCK JUNE 2014

NEUTROPHIL EXTRACELLULAR TRAPS FOLLOWING INJURY

POTENTIAL THERAPEUTIC TARGETS IN NET-RELATED INJURY Reducing NET accumulation in tissues may be important for preventing or attenuating damage to the host. Interfering with NET structure and component proteins could reduce tissue injury following various insults. The potential targets in NET-associated injury are listed in Table 2.

495

combinant human DNase therapy increased circulating IL-6 levels and enhanced sepsis-related tissues damage after cecal ligation and puncture in mice (93). Thammavongsa et al. (94) have reported that Staphylococcus aureus can release nuclease and adenosine synthase during staphylococcal infections in mice, which convert NETs to deoxyadenosine and trigger the caspase 3Ymediated death pathway of immune cells. The ability of S. aureus to cause degradation of NETs is important for the exclusion of macrophages from infection sites (94).

DNase

DNase has a regulatory effect on NET formation in neutrophils and can degrade NETs. DNase levels are increased in early trauma-induced sepsis. Targeting NET components with DNase 1 is effective in protecting mice against TRALI (15). DNase treatment is used for patients with acute lung injury and cystic fibrosis (91). Intrapulmonary DNase is an effective therapy in cystic fibrosis, targeting the extracellular DNA that interferes with mucociliary clearance (91). Antiproteases are also used in therapy to dampen the activity of proteases, and the combined use of antiproteases and DNase may be potentially helpful in controlling NET-mediated lung damage (92). However, previous studies have shown that degradation of NETs may not be a good therapeutic strategy during active infection in a cecalligation-and-puncture mouse model of sepsis (93). The re-

TABLE 2. Therapeutic targets in NET-related injury

Target

Effect on NET formation

Method

References

DNase

j DNase, j proteases

,

Caudrillier et al. (15), Janda and Swiston (91), Greene and McElvaney (92)

Histone

, Histone (antihistone antibody)

,

Caudrillier et al. (15), Martinod et al. (71), Janda and Swiston (91)

,

Li et al. (64), Martinod et al. (71), Knight et al. (97)

PAD4

, PAD4

HMGB1

, HMGB1 (anti-HMGB1 antibody)

,

NADPH oxidase

, NADPH oxidase

,

Hakkim et al. (41), Gray et al. (99)

Gong et al. (76), El (77), Yamada and Maruyama (98)

MPO

, MPO (MPO inhibitor)

,

Narasaraju et al. (16), Parker et al. (100)

SOD

, SOD (SOD inhibitor)

,

Narasaraju et al. (16)

, Platelet function (glycoprotein IIb/IIIa inhibitor, thromboxane A2 inhibitor)

,

Caudrillier et al. (15), et al. (47)

Platelet

NO

, NO

,

Keshari et al. (65), Patel et al. (73)

SP-D

jSP-D

,

Douda et al. (101)

, C5

,

Garcia et al. (102)

C5

SOD indicates superoxide dismutase.

Histones

Histone toxicity originates from the integration of cell membranes, which results in a large inward surge of ion currents and calcium influx (95). Histone-induced NETs could induce surrounding cell death (57). An antihistone antibody may decrease histone-induced cytokine elevation, endothelial damage, coagulation activation, platelet aggregation, and NET formation in a trauma-associated lung injury model (96). The use of antihistone antibodies has been shown to be protective from NET-mediated lung damage (15). Thus, circulating histones are therapeutic targets for improving survival in patients after severe traumatic injury and are considered to be potential drug targets for vessels thrombosis (71). Peptidylarginine deiminase 4

Peptidylarginine deiminase 4 is an important histonemodifying enzyme, and inhibition of PAD4 has been shown to prevent NET formation (64, 97). In addition, histones are modified by PAD4 in the process of NET formation (71). High mobility group box 1

Therapies against HMGB1 have been shown to be beneficial in neutrophil-associated inflammatory conditions, including acute lung injury, sepsis, and IR-induced tissue injury (76, 77, 98). High mobility group box 1 can induce NET formation through a TLR4-dependent pathway and provide a novel therapeutic target for NET-associated injuries (74). NADPH oxidase

Activation of NADPH oxidase is essential for NET formation. Protein kinase C (PKC) plays an important role in NET formation and is a mediator of NADPH oxidase activation (99). Phorbol 12 myristate 13 acetateYinduced NET formation is dependent on ROS production through the activation of NADPH oxidase and PKC (41, 100). Neutrophil extracellular trap formation in response to PMA is dependent on PKC activation, with PKC" being the predominant isoform responsible for NET formation. Inhibition of PKC completely inhibits PMA-induced NET formation (100), suggesting that modulation of NET production via PKC inhibition may offer a novel anti-inflammatory strategy (100). Previous studies have shown that the Raf-MEK-ERK signaling pathway, which is upstream of NADPH oxidase, is critical for NET formation. Drugs that target the Raf-MEK-ERK pathway may also potentially regulate NADPH oxidases and NET formation (41). MPO and superoxide dismutase

Myeloperoxidase activity on NETs could contribute to tissue injury when NETs are released from neutrophils at sites of infection or inflammation (101). Previous studies have shown

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

496

SHOCK VOL. 41, NO. 6

that excessive NET levels can cause alveolar-capillary damage in influenza pneumoniaYinduced acute lung injury. However, following anti-MPO antibody and superoxide dismutase inhibitor treatment, NET formation is decreased, and influenza pneumonitisYinduced lung injury is attenuated (16).

LIU ET

AL.

(b) Neutrophil extracellular traps may serve as a target to develop new therapeutics in the trauma or sepsis in the future. (c) Further clarify the complex mechanisms of NET formation and interactions through various experimental research and clinical studies.

Platelets

Platelet-neutrophil interactions are important for inflammation and hemostasis (75). Previous studies have shown that platelet-induced NET formation is mediated by platelet TLR4 (47). Blockage of platelet-platelet interactions with a glycoprotein IIb/IIIa inhibitor can reduce NET formation and protect against TRALI (15). Accordingly, preventing plateletneutrophil interactions may serve as a novel therapeutic target following injury. Others

Previous studies have shown that NO mediates NET release from human neutrophils through free radical generation. Nitric oxideYmediated formation of NETs is reduced by pretreating neutrophils with the NADPH oxidase inhibitor (diphenyleneiodonium [DPI]) and MPO inhibitor (ABAH: 4-aminobenzoic acid hydrazide) (65, 73). Surfactant protein D (SP-D) is a collectin with innate immune function that is capable of inhibiting NET formation. Therefore, SP-D may serve as another therapeutic candidate for regulating NETosis and NET clearance (101). Garcia et al. (102) suggested that targeting complement 5 (C5) activation may reduce neutrophil infiltration and NET formation in the airways and attenuate influenza virus AYinduced lung injury. Thus, C5 may also be considered as therapeutic target for preventing NET-related injury. CONCLUSIONS Increasing evidence suggests that NETs play an important role in organ function following injury. The pathways involved in NET formation are complex. Reducing NET formation may protect against ischemic brain, limb, and muscle damage, as well as LPS-induced lung injury. Circulating DNase, cf-DNA, and NET levels may serve as markers of trauma and sepsis. Current evidence suggests that efforts in developing potential therapies for pathological NET formation in the setting of trauma or sepsis should focus on targeting DNase, histones, proteases, NADPH oxidase, and MPO. Additional experimental studies and clinical trials are warranted to elucidate the complex mechanisms involved in NET formation, as well as potential therapeutic targets. A better understanding of the underlying mechanisms responsible for the formation of NETs and their effects may provide new insight into how to improve organ function following injury. FUTURE DIRECTIONS In this article, we provide a mini overview regarding the role of NETs in organ injury and offer some perspectives on the future directions for this field. (a) Neutrophil extracellular traps or NETs-related intermediates may be promising biomarkers for the early prediction of organ injury severity in future clinical applications.

REFERENCES 1. Rigby KM, DeLeo FR: Neutrophils in innate host defense against Staphylococcus aureus infections. Semin Immunopathol 34:237Y259, 2012. 2. Kinnula VL, Soini Y, Kvist-Makela K, Savolainen ER, Koistinen P: Antioxidant defense mechanisms in human neutrophils. Antioxid Redox Signal 4:27Y34, 2002. 3. Mayadas TN, Tsokos GC, Tsuboi N: Mechanisms of immune complexYmediated neutrophil recruitment and tissue injury. Circulation 120:2012Y2024, 2009. 4. Schofield ZV, Woodruff TM, Halai R, Wu MC, Cooper MA: NeutrophilsVa key component of ischemia reperfusion injury. Shock 40:463Y470, 2013. 5. Uriarte SM, Rane MJ, Merchant ML, Jin S, Lentsch AB, Ward RA, McLeish KR: Inhibition of neutrophil exocytosis ameliorates acute lung injury in rats. Shock 39:286Y292, 2013. 6. Yu HP, Pang ST, Chaudry IH: Hepatic gene expression patterns following traumahemorrhage: effect of posttreatment with estrogen. Shock 39:77Y82, 2013. 7. Liu FC, Hwang TL, Liu FW, Yu HP: Tropisetron attenuates cardiac injury in a rat trauma-hemorrhage model. Shock 38:76Y81, 2012. 8. Fukudome EY, Li Y, Kochanek AR, Lu J, Smith EJ, Liu B, Kim K, Velmahos GC, deMoya MA, Alam HB: Pharmacologic resuscitation decreases circulating cytokine-induced neutrophil chemoattractant-1 levels and attenuates hemorrhageinduced acute lung injury. Surgery 152:254Y261, 2012. 9. Zhang S, Luo L, Wang Y, Rahman M, Lepsenyi M, Syk I, Jeppsson B, Thorlacius H: Simvastatin protects against T cell immune dysfunction in abdominal sepsis. Shock 38:524Y531, 2012. 10. Land WG, Memer K: Tissue injuryYinduced initiation of the adaptive immune response. Transplantation 93:e38, 2012. 11. Parnell GP, Tang BM, Nalos M, Armstrong NJ, Huang SJ, Booth DR, McLean AS: Identifying key regulatory genes in the whole blood of septic patients to monitor underlying immune dysfunctions. Shock 40:166Y174, 2013. 12. Salomao R, Brunialti MK, Rapozo MM, Baggio-Zappia GL, Galanos C, Freudenberg M: Bacterial sensing, cell signaling, and modulation of the immune response during sepsis. Shock 38:227Y242, 2012. 13. Oklu R, Albadawi H, Jones JE, Yoo HJ, Watkins MT: Reduced hind limb ischemia-reperfusion injury in Toll-like receptor-4 mutant mice is associated with decreased neutrophil extracellular traps. J Vasc Surg 58:1627Y1636, 2013. 14. Simon D, Simon HU, Yousefi S: Extracellular DNA traps in allergic, infectious, and autoimmune diseases. Allergy 68:409Y416, 2013. 15. 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 122:2661Y2671, 2012. 16. Narasaraju T, Yang E, Samy RP, Ng HH, Poh WP, Liew AA, Phoon MC, van RN, Chow VT: Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am J Pathol 179:199Y210, 2011. 17. Zawrotniak M, Rapala-Kozik M: Neutrophil extracellular traps (NETs)Vformation and implications. Acta Biochim Pol 60:277Y284, 2013. 18. Amulic B, Hayes G: Neutrophil extracellular traps. Curr Biol 21:R297YR298, 2011. 19. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A: Neutrophil extracellular traps kill bacteria. Science 303:1532Y1535, 2004. 20. Kaplan MJ, Radic M: Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol 189:2689Y2695, 2012. 21. Thomas GM, Carbo C, Curtis BR, Martinod K, Mazo IB, Schatzberg D, Cifuni SM, Fuchs TA, von Andrian UH, Hartwig JH, et al.: Extracellular DNA traps are associated with the pathogenesis of TRALI in humans and mice. Blood 119:6335Y6343, 2012. 22. Zeng L, Gu W, Zhang AQ, Zhang M, Zhang LY, Du DY, Huang SN, Jiang JX: A functional variant of lipopolysaccharide binding protein predisposes to sepsis and organ dysfunction in patients with major trauma. Ann Surg 255:147Y157, 2012. 23. Herasevich V, Afessa B, Pickering BW: Sepsis in critically ill patients with trauma. Crit Care Med 39:876Y878, 2011. 24. Honda M, Takeichi T, Asonuma K, Tanaka K, Kusunoki M, Inomata Y: Intravital imaging of neutrophil recruitment in hepatic ischemia-reperfusion injury in mice. Transplantation 95:551Y558, 2013. 25. Burdon PC, Martin C, Rankin SM: The CXC chemokine MIP-2 stimulates neutrophil mobilization from the rat bone marrow in a CD49d-dependent manner. Blood 105:2543Y2548, 2005.

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

SHOCK JUNE 2014

NEUTROPHIL EXTRACELLULAR TRAPS FOLLOWING INJURY

26. Singer M, Sansonetti PJ: IL-8 is a key chemokine regulating neutrophil recruitment in a new mouse model of shigella-induced colitis. J Immunol 173:4197Y4206, 2004. 27. Wolters PJ, Wray C, Sutherland RE, Kim SS, Koff J, Mao Y, Frank JA: Neutrophil-derived IL-6 limits alveolar barrier disruption in experimental ventilator-induced lung injury. J Immunol 182:8056Y8062, 2009. 28. Jones MR, Simms BT, Lupa MM, Kogan MS, Mizgerd JP: Lung NF-kappaB activation and neutrophil recruitment require IL-1 and TNF receptor signaling during pneumococcal pneumonia. J Immunol 175:7530Y7535, 2005. 29. 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 8:59Y67, 2002. 30. Visser T, Pillay J, Pickkers P, Leenen LP, Koenderman L: Homology in systemic neutrophil response induced by human experimental endotoxemia and by trauma. Shock 37:145Y151, 2012. 31. Djeu JY, Matsushima K, Oppenheim JJ, Shiotsuki K, Blanchard DK: Functional activation of human neutrophils by recombinant monocyte-derived neutrophil chemotactic factor/IL-8. J Immunol 144:2205Y2210, 1990. 32. Kwak SH, Wang XQ, He Q, Fang WF, Mitra S, Bdeir K, Ploplis VA, Xu Z, Idell S, Cines D, et al.: Plasminogen activator inhibitor-1 potentiates LPSinduced neutrophil activation through a JNK-mediated pathway. Thromb Haemost 95:829Y835, 2006. 33. Kannan S: Role of protease-activated receptors in neutrophil degranulation. Med Hypotheses 59:266Y267, 2002. 34. Bainton DF: Neutrophil granules: a review. Am J Med Technol 42:15Y21, 1976. 35. 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 92:841Y849, 2012. 36. Vethanayagam RR, Almyroudis NG, Grimm MJ, Lewandowski DC, Pham CT, Blackwell TS, Petraitiene R, Petraitis V, Walsh TJ, Urban CF, et al.: Role of NADPH oxidase versus neutrophil proteases in antimicrobial host defense. PLoS One 6:e28149, 2011. 37. Deree J, Lall R, Melbostad H, Grant M, Hoyt DB, Coimbra R: Neutrophil degranulation and the effects of phosphodiesterase inhibition. J Surg Res 133:22Y28, 2006. 38. Arazna M, Pruchniak MP, Demkow U: Neutrophil extracellular traps in bacterial infections: strategies for escaping from killing. Respir Physiol Neurobiol 187:74Y77, 2013. 39. Radic M, Marion TN: Neutrophil extracellular chromatin traps connect innate immune response to autoimmunity. Semin Immunopathol 35:465Y480, 2013. 40. Goldmann O, Medina E: The expanding world of extracellular traps: not only neutrophils but much more. Front Immunol 3:420, 2012. 41. Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zychlinsky A, Waldmann H: Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol 7:75Y77, 2011. 42. Hahn S, Giaglis S, Chowdhury CS, Hosli I, Hasler P: Modulation of neutrophil NETosis: interplay between infectious agents and underlying host physiology. Semin Immunopathol 35:439Y453, 2013. 43. Mesa MA, Vasquez G: NETosis. Autoimmune Dis 2013:651497, 2013. 44. Almyroudis NG, Grimm MJ, Davidson BA, Rohm M, Urban CF, Segal BH: NETosis and NADPH oxidase: at the intersection of host defense, inflammation, and injury. Front Immunol 4:45, 2013. 45. Yipp BG, Kubes P: NETosis: how vital is it? Blood 122:2784Y2794, 2013. 46. Takei H, Araki A, Watanabe H, Ichinose A, Sendo F: Rapid killing of human neutrophils by the potent activator phorbol 12-myristate 13-acetate (PMA) accompanied by changes different from typical apoptosis or necrosis. J Leukoc Biol 59:229Y240, 1996. 47. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, et al.: Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 13:463Y469, 2007. 48. Radic M, Kaplan MJ: Extracellular chromatin traps interconnect cell biology, microbiology, and immunology. Front Immunol 4:160, 2013. 49. Bjornsdottir H, Granfeldt D, Welin A, Bylund J, Karlsson A: Inhibition of phospholipase A(2) abrogates intracellular processing of NADPH-oxidase derived reactive oxygen species in human neutrophils. Exp Cell Res 319:761Y774, 2013. 50. Hamaguchi S, Hirose T, Akeda Y, Matsumoto N, Irisawa T, Seki M, Hosotsubo H, Tasaki O, Oishi K, Shimazu T, et al.: Identification of neutrophil extracellular traps in the blood of patients with systemic inflammatory response syndrome. J Int Med Res 41:162Y168, 2013. 51. Brill A, Fuchs TA, Savchenko AS, Thomas GM, Martinod K, De Meyer SF, Bhandari AA, Wagner DD: Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 10:136Y144, 2012. 52. Palic D, Ostojic J, Andreasen CB, Roth JA: Fish cast NETs: neutrophil extracellular traps are released from fish neutrophils. Dev Comp Immunol 31:805Y816, 2007.

497

53. Cooper PR, Palmer LJ, Chapple IL: Neutrophil extracellular traps as a new paradigm in innate immunity: friend or foe? Periodontol 2000(63):165Y197, 2013. 54. Branzk N, Papayannopoulos V: Molecular mechanisms regulating NETosis in infection and disease. Semin Immunopathol 35:513Y530, 2013. 55. Keshari RS, Jyoti A, Dubey M, Kothari N, Kohli M, Bogra J, Barthwal MK, Dikshit M: Cytokines induced neutrophil extracellular traps formation: implication for the inflammatory disease condition. PLoS One 7:e48111, 2012. 56. Arai Y, Yamashita K, Mizugishi K, Watanabe T, Sakamoto S, Kitano T, Kondo T, Kawabata H, Kadowaki N, Takaori-Kondo A: Serum neutrophil extracellular trap levels predict thrombotic microangiopathy after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 19:1683Y1689, 2013. 57. Saffarzadeh M, Juenemann C, Queisser MA, Lochnit G, Barreto G, Galuska SP, Lohmeyer J, Preissner KT: Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One 7:e32366, 2012. 58. Allen C, Thornton P, Denes A, McColl BW, Pierozynski A, Monestier M, Pinteaux E, Rothwell NJ, Allan SM: Neutrophil cerebrovascular transmigration triggers rapid neurotoxicity through release of proteases associated with decondensed DNA. J Immunol 189:381Y392, 2012. 59. Cao CX, Yang QW, Lv FL, Cui J, Fu HB, Wang JZ: Reduced cerebral ischemia-reperfusion injury in Toll-like receptor 4 deficient mice. Biochem Biophys Res Commun 353:509Y514, 2007. 60. Enzmann G, Mysiorek C, Gorina R, Cheng YJ, Ghavampour S, Hannocks MJ, Prinz V, Dirnagl U, Endres M, Prinz M, et al.: The neurovascular unit as a selective barrier to polymorphonuclear granulocyte (PMN) infiltration into the brain after ischemic injury. Acta Neuropathol 125:395Y412, 2013. 61. Doring Y, Weber C, Soehnlein O: Footprints of neutrophil extracellular traps as predictors of cardiovascular risk. Arterioscler Thromb Vasc Biol 33: 1735Y1736, 2013. 62. McDonald B, Urrutia R, Yipp BG, Jenne CN, Kubes P: Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis. Cell Host Microbe 12:324Y333, 2012. 63. Fuchs TA, Brill A, Wagner DD: Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 32:1777Y1783, 2012. 64. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y: PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med 207:1853Y1862, 2010. 65. Keshari RS, Jyoti A, Kumar S, Dubey M, Verma A, Srinag BS, Krishnamurthy H, Barthwal MK, Dikshit M: Neutrophil extracellular traps contain mitochondrial as well as nuclear DNA and exhibit inflammatory potential. Cytometry A 81:238Y247, 2012. 66. Tadie JM, Bae HB, Jiang S, Park DW, Bell CP, Yang H, Pittet JF, Tracey K, Thannickal VJ, Abraham E, et al.: HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Physiol Lung Cell Mol Physiol 304:L342YL349, 2013. 67. Itakura A, McCarty OJ: Pivotal role for the mTOR pathway in the formation of neutrophil extracellular traps via regulation of autophagy. Am J Physiol Cell Physiol 305:C348YC354, 2013. 68. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA, et al.: Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 184:205Y213, 2009. 69. Neeli I, Khan SN, Radic M: Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol 180:1895Y1902, 2008. 70. Neeli I, Dwivedi N, Khan S, Radic M: Regulation of extracellular chromatin release from neutrophils. J Innate Immun 1:194Y201, 2009. 71. Martinod K, Demers M, Fuchs TA, Wong SL, Brill A, Gallant M, Hu J, Wang Y, Wagner DD: Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A 110:8674Y8679, 2013. 72. Pilsczek FH, Salina D, Poon KK, Fahey C, Yipp BG, Sibley CD, Robbins SM, Green FH, Surette MG, Sugai M, et al.: A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol 185:7413Y7425, 2010. 73. Patel S, Kumar S, Jyoti A, Srinag BS, Keshari RS, Saluja R, Verma A, Mitra K, Barthwal MK, Krishnamurthy H, et al.: Nitric oxide donors release extracellular traps from human neutrophils by augmenting free radical generation. Nitric Oxide 22:226Y234, 2010. 74. Tadie JM, Bae HB, Jiang S, Park DW, Bell CP, Yang H, Pittet JF, Tracey K, Thannickal VJ, Abraham E, et al.: HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Physiol Lung Cell Mol Physiol 304:L342YL349, 2013. 75. Ghasemzadeh M, Hosseini E: Platelet-leukocyte crosstalk: linking proinflammatory responses to procoagulant state. Thromb Res 131:191Y197, 2013. 76. Gong G, Yuan LB, Hu L, Wu W, Yin L, Hou JL, Liu YH, Zhou LS: Glycyrrhizin attenuates rat ischemic spinal cord injury by suppressing inflammatory cytokines and HMGB1. Acta Pharmacol Sin 33:11Y18, 2012.

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

498

SHOCK VOL. 41, NO. 6

77. El GM: HMGB1 modulates inflammatory responses in LPS-activated macrophages. Inflamm Res 56:162Y167, 2007. 78. Tadie JM, Bae HB, Banerjee S, Zmijewski JW, Abraham E: Differential activation of RAGE by HMGB1 modulates neutrophil-associated NADPH oxidase activity and bacterial killing. Am J Physiol Cell Physiol 302:C249YC256, 2012. 79. Itakura A, McCarty OJ: Pivotal role for the mTOR pathway in the formation of neutrophil extracellular traps via regulation of autophagy. Am J Physiol Cell Physiol 305:C348YC354, 2013. 80. McInturff AM, Cody MJ, Elliott EA, Glenn JW, Rowley JW, Rondina MT, Yost CC: Mammalian target of rapamycin regulates neutrophil extracellular trap formation via induction of hypoxia-inducible factor 1 alpha. Blood 120:3118Y3125, 2012. 81. Meng W, Paunel-Gorgulu A, Flohe S, Witte I, Schadel-Hopfner M, Windolf J, Logters TT: Deoxyribonuclease is a potential counter regulator of aberrant neutrophil extracellular traps formation after major trauma. Mediators Inflamm 2012:149560, 2012. 82. Logters T, Paunel-Gorgulu A, Zilkens C, Altrichter J, Scholz M, Thelen S, Krauspe R, Margraf S, Jeri T, Windolf J, et al.: Diagnostic accuracy of neutrophil-derived circulating free DNA (cf-DNA/NETs) for septic arthritis. J Orthop Res 27:1401Y1407, 2009. 83. Margraf S, Logters T, Reipen J, Altrichter J, Scholz M, Windolf J: Neutrophilderived circulating free DNA (cf-DNA/NETs): a potential prognostic marker for posttraumatic development of inflammatory second hit and sepsis. Shock 30:352Y358, 2008. 84. Breitbach S, Tug S, Simon P: Circulating cell-free DNA: an up-coming molecular marker in exercise physiology. Sports Med 42:565Y586, 2012. 85. Napirei M, Ricken A, Eulitz D, Knoop H, Mannherz HG: Expression pattern of the deoxyribonuclease 1 gene: lessons from the Dnase1 knockout mouse. Biochem J 380:929Y937, 2004. 86. Jaillon S, Peri G, Delneste Y, Fremaux I, Doni A, Moalli F, Garlanda C, Romani L, Gascan H, Bellocchio S, et al.: The humoral pattern recognition receptor PTX3 is stored in neutrophil granules and localizes in extracellular traps. J Exp Med 204:793Y804, 2007. 87. Deban L, Russo RC, Sironi M, Moalli F, Scanziani M, Zambelli V, Cuccovillo I, Bastone A, Gobbi M, Valentino S, et al.: Regulation of leukocyte recruitment by the long pentraxin PTX3. Nat Immunol 11:328Y334, 2010. 88. Bottazzi B, Garlanda C, Cotena A, Moalli F, Jaillon S, Deban L, Mantovani A: The long pentraxin PTX3 as a prototypic humoral pattern recognition receptor: interplay with cellular innate immunity. Immunol Rev 227:9Y18, 2009. 89. Daigo K, Hamakubo T: Host-protective effect of circulating pentraxin 3 (PTX3) and complex formation with neutrophil extracellular traps. Front Immunol 3:378, 2012.

LIU ET

AL.

90. Savchenko AS, Inoue A, Ohashi R, Jiang S, Hasegawa G, Tanaka T, Hamakubo T, Kodama T, Aoyagi Y, Ushiki T, et al.: Long pentraxin 3 (PTX3) expression and release by neutrophils in vitro and in ulcerative colitis. Pathol Int 61:290Y297, 2011. 91. Janda S, Swiston J: Intrapleural fibrinolytic therapy for treatment of adult parapneumonic effusions and empyemas: a systematic review and meta-analysis. Chest 142:401Y411, 2012. 92. Greene CM, McElvaney NG: Proteases and antiproteases in chronic neutrophilic lung diseaseVrelevance to drug discovery. Br J Pharmacol 158:1048Y1058, 2009. 93. Meng W, Paunel-Gorgulu A, Flohe S, Hoffmann A, Witte I, Mackenzie C, Baldus SE, Windolf J, Logters TT: Depletion of neutrophil extracellular traps in vivo results in hypersusceptibility to polymicrobial sepsis in mice. Crit Care 16:R137, 2012. 94. Thammavongsa V, Missiakas DM, Schneewind O: Staphylococcus aureus degrades neutrophil extracellular traps to promote immune cell death. Science 342:863Y866, 2013. 95. Waring P: Redox active calcium ion channels and cell death. Arch Biochem Biophys 434:33Y42, 2005. 96. Abrams ST, Zhang N, Manson J, Liu T, Dart C, Baluwa F, Wang SS, Brohi K, Kipar A, Yu W, et al.: Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med 187:160Y169, 2013. 97. Knight JS, Zhao W, Luo W, Subramanian V, O’Dell AA, Yalavarthi S, Hodgin JB, Eitzman DT, Thompson PR, Kaplan MJ: Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J Clin Invest 123:2981Y2993, 2013. 98. Yamada S, Maruyama I: HMGB1, a novel inflammatory cytokine. Clin Chim Acta 375:36Y42, 2007. 99. Gray RD, Lucas CD, Mackellar A, Li F, Hiersemenzel K, Haslett C, Davidson DJ, Rossi AG: Activation of conventional protein kinase C (PKC) is critical in the generation of human neutrophil extracellular traps. J Inflamm (Lond) 10:12, 2013. 100. Parker H, Albrett AM, Kettle AJ, Winterbourn CC: Myeloperoxidase associated with neutrophil extracellular traps is active and mediates bacterial killing in the presence of hydrogen peroxide. J Leukoc Biol 91:369Y376, 2012. 101. 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: 1856Y1865, 2011. 102. Garcia CC, Weston-Davies W, Russo RC, Tavares LP, Rachid MA, ves-Filho JC, Machado AV, Ryffel B, Nunn MA, Teixeira MM: Complement C5 activation during influenza A infection in mice contributes to neutrophil recruitment and lung injury. PLoS One 8:e64443, 2013.

Copyright © 2014 by the Shock Society. Unauthorized reproduction of this article is prohibited.

Role of neutrophil extracellular traps following injury.

Neutrophil extracellular traps (NETs), which consist of neutrophil DNA and cytoplasmic proteins, have been shown to be involved in various infectious,...
3MB Sizes 6 Downloads 5 Views