Skip main navigation

Neutrophil Extracellular Traps Promote Thrombin Generation Through Platelet-Dependent and Platelet-Independent Mechanisms

Originally publishedhttps://doi.org/10.1161/ATVBAHA.114.304114Arteriosclerosis, Thrombosis, and Vascular Biology. 2014;34:1977–1984

Abstract

Objective—

Activation of neutrophils by microbial or inflammatory stimuli results in the release of neutrophil extracellular traps (NETs) that are composed of DNA, histones, and antimicrobial proteins. In purified systems, cell-free DNA (CFDNA) activates the intrinsic pathway of coagulation, whereas histones promote thrombin generation through platelet-dependent mechanisms. However, the overall procoagulant effects of CFDNA/histone complexes as part of intact NETs are unknown. In this study, we examined the procoagulant potential of intact NETs released from activated neutrophils. We also determined the relative contribution of CFDNA and histones to thrombin generation in plasmas from patients with sepsis.

Approach and Results—

NETs released from phorbyl myristate–activated neutrophils enhance thrombin generation in platelet-poor plasma. This effect was DNA dependent (confirmed by DNase treatment) and occurred via the intrinsic pathway of coagulation (confirmed with coagulation factor XII– and coagulation factor XI–depleted plasma). In platelet-rich plasma treated with corn trypsin inhibitor, addition of phorbyl myristate–activated neutrophils increased thrombin generation and shortened the lag time in a toll-like receptor-2– and toll-like receptor-4–dependent mechanism. Addition of DNase further augmented thrombin generation, suggesting that dismantling of the NET scaffold increases histone-mediated, platelet-dependent thrombin generation. In platelet-poor plasma samples from patients with sepsis, we found a positive correlation between endogenous CFDNA and thrombin generation, and addition of DNase attenuated thrombin generation.

Conclusions—

These studies examine the procoagulant activities of CFDNA and histones in the context of NETs. Our studies also implicate a role for the intrinsic pathway of coagulation in sepsis pathogenesis.

Introduction

Sepsis is the leading cause of morbidity and mortality in noncoronary intensive care units in the Western world.1 Severe sepsis, defined as sepsis associated with ≥1 dysfunctional organ, afflicts ≈750 000 individuals in the United States annually, with an estimated mortality rate of 30% to 50%.1 Sepsis is often initiated by release of microorganisms and/or microbial toxins into the circulation.2,3 Under these conditions, morbidity and mortality are the result of uncontrolled activation of inflammatory and coagulation pathways, which leads to microvascular thrombosis and subsequent multiple organ dysfunction syndrome.

Although many clinical trials have explored the use of agents designed to attenuate inflammatory and/or coagulation pathways, all have failed, and the outcome of patients with severe sepsis remains poor.4,5 Thus, a better understanding of the pathogenesis of sepsis is needed. Recently, cell-free DNA (CFDNA) has emerged as an important link among innate immunity, coagulation, and inflammation.68 When activated by microbial or inflammatory stimuli, neutrophils release web-like structures known as neutrophil extracellular traps (NETs), which are composed of CFDNA, histones, and antimicrobial proteins.7 These structures bind to microorganisms, prevent them from spreading, and ensure a high local concentration of neutrophil granule enzymes to kill bacteria.

CFDNA is the major structural component of NETs, as shown by the ability of DNA-intercalating dyes to stain NETs and by the ability of DNase to dismantle NETs.7 However, CFDNA might also have deleterious effects on the host. CFDNA triggers the intrinsic pathway of blood coagulation,9 and elevated levels of CFDNA are found in patients with deep vein thrombosis.8,10 It has been proposed that the presence of CFDNA and platelet–neutrophil interactions in the microcirculation results in microvascular thrombosis, leading to tissue hypoxia and endothelial damage.11,12

Histones, the other principal component of extracellular traps, are important contributors to the bactericidal and cytotoxic properties of NETs.13 Histones are cationic nuclear proteins that associate with DNA to form nucleosomes, the repeating units of chromatin. When injected into mice, histones result in death because of an extreme prothrombotic response, including diffuse microvascular thrombosis, fibrin deposition, platelet aggregation, and thrombocytopenia.14 Histone H4 is cytotoxic toward endothelial cells, and blocking histone-mediated cytotoxicity protects mice from endotoxemia.14 In purified systems, histones H3 and H4 directly induce platelet aggregation through interactions with toll-like receptors (TLRs) 2 and 4.15

Many of the studies on the procoagulant/proinflammatory properties of CFDNA and histone proteins have examined these components in isolation. However, the majority of CFDNA in plasma is likely histone-bound. Thus, the overall procoagulant effects of CFDNA/histone complexes as part of intact NETs released from activated neutrophils are unknown. Importantly, the interaction between CFDNA and histones may shield many of the pathophysiological effects observed when components are examined in isolation. To address this possibility, we (1) identified the cells responsible for release of CFDNA in blood, (2) compared the capacity of NETs released from activated neutrophils to promote thrombin generation in platelet-poor plasma (PPP) and platelet-rich plasma (PRP), and (3) determined the relative contribution of CFDNA and histones to thrombin generation in plasma from patients with sepsis.

Materials and Methods

The study design and experimental methods are described in detail in the online-only Supplement.

Results

Neutrophils Are the Major Source of Plasma CFDNA Released From Activated Whole Blood

Previously, we reported that high levels of CFDNA in plasma predicts poor clinical outcome in patients with severe sepsis.16 DNA sequence analyses and studies with TLR9 reporter cells suggest that the circulating CFDNA from patients with sepsis is host derived.16 In this study, we determined whether neutrophils are the major source of circulating CFDNA released from activated whole blood. Incubation of whole blood with lipopolysaccharide (Figure 1A) or lipoteichoic acid and peptidoglycan (Figure 1B) produced a rapid increase in plasma CFDNA. Similarly, incubation of purified neutrophils with lipopolysaccharide (Figure 1C) or lipoteichoic acid/peptidoglycan (Figure 1D) also resulted in a rapid increase in plasma CFDNA. The amount of CFDNA released by activated neutrophils was similar to that released by activated whole blood, suggesting that neutrophils are the primary source of CFDNA when whole blood is incubated with lipopolysaccharide or lipoteichoic acid+peptidoglycan.

Figure 1.

Figure 1. Cell-free DNA (CFDNA) release from activated neutrophils. Human whole blood was stimulated for varying lengths of time with increasing concentrations of lipopolysaccharide (LPS; A) or lipoteichoic acid (LTA)/peptidoglycan (B), and levels of CFDNA were measured. Isolated human neutrophils were treated with increasing doses of LPS (C) or LTA/peptidoglycan, and levels of CFDNA were measured (n=3 independent experiments). *P<0.05 indicates significance relative to untreated conditions.

To confirm that the increase in CFDNA from activated whole blood or neutrophils was because of the release of NETs, we performed the studies in the presence of Cl-amidine, an inhibitor of NET formation.17,18 Specifically, Cl-amidine inhibits peptidyl arginine deiminase type IV deimination activity by covalently modifying an active site cysteine on peptidyl arginine deiminase type IV required for chromatin decondensation. As shown in Figure 1, the inclusion of Cl-amidine abrogated the release of CFDNA in whole blood, as well as in purified neutrophils, confirming that the increase in CFDNA is because of the release of NETs. To visualize the released CFDNA, neutrophils mounted on coverslips were imaged after stimulation with increasing concentrations of phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide, and lipoteichoic acid/peptidoglycan. As shown in Figure 2A and Figure II in the online-only Data Supplement, the release of extracellular chromatin was observed in neutrophils stimulated with PMA and bacterial components but not in resting neutrophils.

Figure 2.

Figure 2. Phorbol-12-myristate-13-acetate (PMA)–stimulated neutrophils released neutrophil extracellular traps (NETs) that enhance thrombin generation via the intrinsic pathway. Human neutrophils were incubated without (top) or with (bottom) 100 nmol/L PMA for 30 minutes, and both intra- and extracellular DNA was stained with DAPI (4’,6-diamidino-2-phenylindole; blue) and Sytox Green (green), respectively (A). The effects of NETs on thrombin generation were examined as described in Materials and Methods (B). The effects of NETs on thrombin generation were also measured in coagulation factor XI (FXI)– (C) and FXII-deficient (D) plasma. A summary of coagulation parameters for NET release in normal plasma is described in Table 1. Thrombograms shown are representative of 4 to 6 independent experiments. *P<0.05 indicates significance relative to plasma only conditions. FXIIdp indicates coagulation factor XII–deficient plasma; and PPP, platelet-poor plasma.

NETs Enhance Thrombin Generation Through the Intrinsic Pathway

To date, there have been no studies that have examined plasma thrombin generation in the presence of intact NETs released from activated neutrophils. To study the effects of NETs on thrombin generation, neutrophils isolated from healthy volunteers were added to PPP and the plasma was then incubated for 30 minutes in the absence or presence of PMA before recalcification and quantification of thrombin generation. In the presence of PMA, the lag time and time to peak thrombin were shorter, and peak thrombin and total thrombin (area under the curve) were higher compared with those measured in the absence of PMA, findings consistent with a procoagulant effect (Figure 2B; Table 1; Table I in the online-only Data Supplement). This activity was diminished with DNase I but not with RNase (data not shown), findings that suggest that the procoagulant activity is mediated by CFDNA.

Table 1. Effect of NET Release on Thrombin Generation in Platelet-Poor Plasma

Lag Time, min Peak IIa, nmol/L Peak Time, min AUC
PPP only 28±4 58±17 41±5 1188±375
Neutrophils 24±2 62±6 42±4 1375±140
NETs 13±1* 225±23* 20±2* 2898±383*

Summary of coagulation parameters for NET release in corn trypsin inhibitor–supplemented PPP. Results reflect the mean±SE of ≥3 determinations. AUC indicates area under the curve; NET, neutrophil extracellular trap; and PPP, platelet-poor plasma.

*P<0.05 compared with PPP.

Although similar results were obtained in coagulation factor VII (FVII)–deficient plasma, when thrombin generation was quantified in FXII- or FXI-deficient plasma, incubation with PMA-activated neutrophils had little effect. The results suggest that the procoagulant activity of CFDNA is mediated via the intrinsic pathway. This concept is supported by the observations that (1) supplementation of FXII- or FXI-deficient plasma with FXII and FXI, respectively, restores procoagulant activity (Figure 2C and 2D), and (2) addition of corn trypsin inhibitor (CTI), a potent and specific inhibitor of FXIIa, to control plasma abolishes procoagulant activity (Figure 2).

NETs Enhance Thrombin Generation in a Platelet-Dependent Manner

Purified histones have been reported to enhance thrombin generation in PRP through a polyphosphate-dependent mechanism.19 However, it remains unclear whether the platelet-activating effects of histones are shielded when in complex with CFDNA and other NET components. To determine whether platelets enhance thrombin generation when neutrophil-containing plasma is incubated with PMA, results in PRP were compared with those in PPP. We induced thrombin generation in CTI-inhibited PRP in the absence or presence of PMA-activated neutrophils. In this system, CTI prevents CFDNA-mediated FXII activation and subsequent thrombin generation but is unable to inhibit platelet-mediated (polyphosphate-dependent) FXII activation.19 In the presence of neutrophils, incubation of PRP with PMA shortened the lag time by half and increased peak thrombin compared with the lag time and peak thrombin determined in PPP. The addition of PMA to PRP caused a modest increase in thrombin generation (consistent with the known platelet-activating effects of PMA),20 whereas the addition of PMA-activated neutrophils significantly exacerbated this effect. Compared with PMA-treated PRP, the addition of PMA-activated neutrophils to PRP resulted in a 50% decrease in lag time, accompanied by a significant increase in peak and total thrombin (Figure 3; Table 2). The enhanced procoagulant effect was attenuated with bovine alkaline phosphatase (AP), suggesting that it is mediated by inorganic polyphosphate released from platelets. Thrombin generation in PRP was further enhanced with DNase addition but not with RNase addition. These findings suggest that dismantling of the DNA network of NETs with DNase releases more procoagulant material (presumably histones; Table 2). Addition of TLR2- and/or TLR4-directed inhibitory antibodies attenuated this enhancement (Figure 4A, B), whereas tissue factor–inhibitory antibody HTF-1 or a control IgG had no effect (Table 2). Taken together, these findings suggest that digestion of the DNA network of NETs with DNase exposes the platelet-activating functions of histones.

Table 2. Effect of NET Release on Thrombin Generation in PRP

Lag Time, min Peak IIa, nmol/L Peak Time, min AUC
DNase + + + +
PRP 76±2 81±3 63±1 109±11 89±2 101±4 2730±224 2648±378
Neutrophils 73±1 69±8 85±3 77±16 94±2 86±7 2695±243 2952±721
PMA 61±16 59±6 101±9 97±12 73±6 74±11 2910±510 2848±193
NETs 37±2* 32±2* 203±13* 234±15* 46±2* 40±2* 3608±324* 3631±329*
NET+Cl-amidine 65±12 72±8 68±9 88±5 79±7 86±13 2485±588 2544±431
NET+TLR2 41±1 45±9 133±6 161±7 48±1 53±2 3219±594 3048±203
NET+TLR4 46±2 47±10 174±8 168±4 54±4 56±5 3831±1076 3578±359
NET+TLR2 and 4 51±2 56±8 140±14 153±11 59±1 69±7 3644±1117 2956±520
NET+H3 mAb 44±7 52±3 177±5 161±3 55±4 63±3 3375±643 3058±244
NET+H4 mAb 46±2 55±4 172±5 132±14 57±6 66±2 3687±385 3184±195
NET+H3+H4 mAbs 49±3 58±12 143±9 101±10 61±4 64±6 3298±343 2944±318
NET+HTF1 39±2 31±1 210±6 253±13 47±3 39±5 3918±341 4032±412
NET+IgG 35±2 40±6 202±18 178±8 44±4 51±6 5226±278 4772±821
NET+AP 89±2 91±3 3±1 8±4 111±6 98±11 22±14 46±18

Summary of coagulation parameters for NET release in corn trypsin inhibitor–supplemented PRP. Results reflect the mean±SE of ≥3 determinations. AP indicates alkaline phosphatase; AUC, area under the curve; mAb, monoclonal antibody; NET, neutrophil extracellular trap; PMA, phorbol-12-myristate-13-acetate; PRP, platelet-rich plasma; and TLR, toll-like receptor.

*P<0.05 compared with unstimulated neutrophils.

P<0.05 compared with NETting neutrophils.

Figure 3.

Figure 3. Effect of intact neutrophil extracellular traps (NETs) on platelet-mediated thrombin generation. Thrombin generation induced by NETting neutrophils in corn trypsin inhibitor–inhibited platelet-rich plasma (PRP) with or without deoxyribonuclease I (DNase I; 20 μg/mL) treatment to degrade NET structures. Thrombograms shown are representative of 3 independent experiments. A summary of coagulation parameters for NET release in normal plasma is described in Table 2. Thrombograms shown are representative of 4 independent experiments. *P<0.05 indicates significance relative to unstimulated neutrophil conditions. PMA indicates phorbol-12-myristate-13-acetate.

Figure 4.

Figure 4. Neutrophil extracellular trap (NET)–induced platelet activation is mediated by extracellular histones through toll-like receptor (TLR) 2 and TLR4. A, Thrombin generation induced by intact NETs in corn trypsin inhibitor (CTI)–inhibited platelet-rich plasma (PRP) with inhibitory monoclonal antibodies (mAbs) against TLR2, TLR4, or bovine alkaline phosphatase (AP). B, Thrombin generation induced by deoxyribonuclease (DNase)-digested NETs in CTI-inhibited PRP with inhibitory antibodies against TLR2/4 and inhibitory antibodies to histones H3 and H4. Thrombograms shown are representative of 3 independent experiments. A summary of coagulation parameters for NET release in CTI-supplemented PRP is described in Table 3. *P<0.05 indicates significance relative to NETting neutrophil–only conditions.

DNA–Histone Complexes in Plasma From Patients With Severe Sepsis Enhance Thrombin Generation

To determine the physiological relevance of our in vitro studies, we measured plasma levels of DNA–histone complexes in patients with sepsis. Compared with plasma from healthy controls, plasma from patients with severe sepsis contained increased levels of DNA–histone complexes (Figure 5), suggesting that CFDNA circulates in complex with histones. Next, we investigated whether there is a correlation between plasma CFDNA levels and plasma thrombin generation. Plasma samples from severe sepsis patients were divided into those that contained low, intermediate, or high levels of CFDNA as arbitrarily defined as CFDNA levels <5 μg mL−1, 5.0 to 14.9 μg mL−1, and >15 μg mL−1, respectively. Thrombin generation in these samples was then determined in the absence or presence of DNase. In the absence of DNase, there was a direct correlation between CFDNA levels and total thrombin as determined by area under the curve (r=0.6; Figure 6F) and an inverse correlation between CFDNA levels and lag times (r=0.56; Figure 6G). Similarly, higher CFDNA levels were associated with shorter lag times (Figure 6B) and times to peak (Figure 6C), higher peak thrombin values (Figure 6D), as well as greater area under the curve (Figure 6E) relative to control. Incubation with DNase (confirmed by gel electrophoresis; data not shown) attenuated thrombin generation in septic plasma and DNase addition to control plasma-reduced thrombin generation to undetectable levels (Figure 6). The addition of protamine sulfate, a small cationic protein that binds and precipitates DNA, also reduced thrombin generation to undetectable levels (Figure 6A). In addition, although CTI inclusion resulted in an abrogation of thrombin generation, no effect was seen when tissue factor–inhibitory antibody HTF-1 was added to septic plasmas, suggesting negligible contributions by tissue factor to this system (Table 3). Taken together, these data suggest that elevations in plasma CFDNA levels result in a hypercoagulable state in patients with sepsis.

Table 3. Effect of Increasing Concentrations of CFDNA on Thrombin Generation in Plasmas of Patients With Sepsis

Lag Time, min Peak IIa, nmol/L Peak Time, min AUC
Normal 27±2 166±32 34±2 2592±326
Low [CFDNA] 17±2* 362±53* 20±2* 3189±326*
Intermediate [CFDNA] 14±2* 446±12* 17±3* 3456±133*
High [CFDNA] 11±1* 480±12* 14±1* 3697±139*
High [CFDNA]+DNase I 0 0 0 0
High [CFDNA]+HTF-1 12±3* 476±18* 14±2* 3702±267*
High [CFDNA]+CTI 0 0 0 0

Summary of coagulation parameters for thrombin generation in septic patient plasmas. Results reflect the mean±SE of 5 determinations. AUC indicates area under the curve; CFDNA, cell-free DNA; CTI, corn trypsin inhibitor; and DNase, deoxyribonuclease.

*P<0.05 relative to normal plasma.

Figure 5.

Figure 5. Increased levels of nucleosomes correspond with increases in cell-free DNA (CFDNA) in sepsis. CFDNA was isolated from the plasma of healthy volunteers and patients with sepsis (A). Concentrations of circulating nucleosomes were determined using the Cell Death Detection ELISA PLUS from Roche Diagnostics (B; n=10 for each group).

Figure 6.

Figure 6. Effects of elevated levels of cell-free DNA (CFDNA) in septic plasmas on thrombin generation. Plasmas obtained from patients with severe sepsis were recalcified to initiate coagulation, and thrombin generation was measured as described in Materials and Methods (A). Patients were categorized based on CFDNA concentrations: low CFDNA levels () <5 μg/mL; intermediate CFDNA levels (▼) 5.1 to 14.9 μg/mL, and high CFDNA levels (Δ) >15 μg/mL. Deoxyribonuclease (+/−) indicates the presence or absence of a 4-hour deoxyribonuclease (DNase) pretreatment before initiating thrombin generation. Lag time (B), time to peak (C), peak thrombin (D), and area under the curve (AUC; E) analysis was performed with Technothrombin TGA software. Correlation curves for AUC and CFDNA levels (F) as well as lag time and CFDNA levels (G). *P<0.05 and **P<0.01 indicate significance relative to plasma only. Thrombograms shown are representative of 5 independent experiments (n=5 for all subgroups). Ctrl indicates control; ND, no thrombin generation detected; and PS, protamine sulfate.

Discussion

CFDNAs are extracellular DNA fragments that circulate within peripheral blood. CFDNA circulates at low levels in healthy individuals, with elevated levels observed in an array of clinical conditions including trauma,21 cancer,9 stroke,22 myocardial infarction,23 and sepsis.24,25 In a previous study, we demonstrated that CFDNA seems to have high discriminative power to predict intensive care unit mortality in patients with severe sepsis.16 Patients with higher plasma concentrations of CFDNA are more likely to face severe complications, such as organ dysfunction/failure and death. As a prognostic indicator of mortality in septic intensive care unit patients, CFDNA alone seems to possess the greatest predictive power, even when combined with existing clinical scoring systems such as MODS (Multiple Organ Dysfunction Score) and APACHE II (Acute Physiology and Chronic Health Evaluation II) scores.16

Our present work reveals 4 major findings. First, we identified neutrophil-derived NETs as the most likely source of elevated CFDNA in whole blood exposed to microbial toxins. Second, we demonstrated that intact NETs promote thrombin generation in PPP and that (1) thrombin generation is triggered via the intrinsic pathway, and (2) thrombin generation in PPP is attenuated with DNase I but not RNase. Previous studies have focused on purified NET components or NETs that had been processed before use and thus may lack a physiologically relevant structural integrity.19,26 Third, we demonstrated that dismantling the NET scaffold with DNase increases histone-mediated, platelet-dependent thrombin generation. This observation may explain why administration of DNase in septic mice results in organ damage and decreased survival and warrants future studies to explore the therapeutic effects of antihistone therapy in experimental sepsis.27,28 Finally, our work demonstrates elevations in indices of thrombin generation in plasma samples from patients with sepsis and implicate an important role of the intrinsic pathway of coagulation in the pathogenesis of sepsis.

For many years, the hemostatic abnormalities in sepsis have been described as an initial hypercoagulable phase driven by aberrant expression of tissue factor, downregulation of endogenous anticoagulant pathways, and impairment of fibrinolysis because of elevations in plasminogen activator inhibitor-1.24 Currently, the thrombin generation assay is one of the most extensively used global hemostatic assays used in hemostasis research.29 However, several groups have reported that patients with sepsis present no signs of systemic hypercoagulability when evaluated with the thrombin generation assay, even in the early stages of sepsis.3034 It should be noted that thrombin generation in previous studies was analyzed in PPP triggered with relipidated tissue factor (ie, via the extrinsic pathway) rather than with CaCl2 as was done in the present study. Thus, previous studies have overlooked the importance of the intrinsic pathway of blood coagulation in the hypercoagulable state observed in patients with sepsis.

Given that CFDNA is a potent activator of coagulation, lowering levels of CFDNA may be beneficial to the host in disease states. In humans and mouse models, there is precedence for the therapeutic efficacy of DNase. For example, in patients with cystic fibrosis, a condition often associated with Pseudomonas aeruginosa infection of the lung epithelium, inhalation of recombinant human DNase I reduces the viscosity of purulent sputum and inhibits bacterial biofilm formation.35 In a mouse model of systemic lupus erythematosus, an autoimmune disease characterized by high circulating DNA levels, intraperitoneal injection of recombinant mouse DNase interferes with the disease process.36 In the present study, however, we have observed that addition of DNase to NETs actually results in increases in thrombin generation in PRP, suggesting that removal of the CFDNA component of NETs may be detrimental to the host. Consistent with this finding, Meng et al27 showed that early digestion of NETs by DNase in a mouse model of sepsis results in advanced sepsis progression accompanied by an increase in mortality. It is possible that, in addition to impairing bactericidal capabilities, early digestion of NETs with DNase exposes histones that are potent activators of platelets and are cytotoxic to vascular endothelial cells. Thus, future studies should aim to better understand the therapeutic potential of degrading NETs in sepsis.

Because histones are presumably exposed when the CFDNA scaffold is degraded by DNase, the absence of thrombin generation in PPP incubated with PMA/DNase-treated neutrophils or purified histones suggests that histones themselves do not trigger the intrinsic pathway of coagulation. It also suggests that neutrophil granular enzymes do not trigger the intrinsic pathway. However, it is possible that neutrophil enzymes promote coagulation by inactivating endogenous anticoagulants. Neutrophil elastase has been shown to degrade antithrombin, and both elastase and cathespin G (both released during neutrophil degranulation) proteolyze tissue factor pathway inhibitor.37,38

Semeraro et al19 have recently demonstrated that purified histones activate platelets through TLR2 and TLR4, inducing the secretion of inorganic polymer polyP. When thrombin generation was performed in the presence of NETs in CTI-treated PRP (which inhibits CFDNA-mediated but not polyphosphate-mediated contact activation), there was a robust reduction in lag time coupled with an increase in peak thrombin, suggesting that, similar to purified histone proteins, intact NETs are able to activate platelets. This effect was platelet dependent because no thrombin generation was detected when CTI-inhibited PPP was used. However, platelet activation in the presence of NETs was not completely abrogated with TLR2- and TLR4-blocking antibodies, suggesting that alternative mechanisms may also regulate platelet activation in the presence of NETs. Histones have been shown to increase membrane permeability of cells, which contribute to their cytotoxic effects.14,39

In addition to histones, platelet polyP may be another therapeutic target in sepsis. Recent studies have suggested a therapeutic use for AP in the treatment of sepsis-associated organ dysfunction.40 Two phase II studies demonstrated that parenteral administration of the dephosphorylating enzyme AP to intensive care unit patients with sepsis and associated acute kidney injury improved kidney function and reduced markers of inflammation and kidney injury.41,42 The therapeutic efficacy of AP has been attributed to AP-mediated dephosphorylation/detoxification of lipopolysaccharide and dephosphorylation of ATP, a proinflammatory energy molecule released by inflamed renal tissue.40 In the present study, we have shown that the addition of AP abolished thrombin generation in PRP, suggesting that AP may also exert beneficial effects by impairing platelet polyP-dependent activation of coagulation.

In summary, these studies examine the procoagulant activities of CFDNA and histones in the context of intact NETs. Our studies also implicate a role for CFDNA-mediated activation of the intrinsic pathway of coagulation in the pathogenesis of sepsis. Our findings support the concept that NET components may be important therapeutic targets for the treatment of sepsis

Nonstandard Abbreviations and Acronyms

AP

alkaline phosphatase

CFDNA

cell-free DNA

CTI

corn trypsin inhibitor

FVII/FXI/FXII

coagulation factor VII/XI/XII

NETS

neutrophil extracellular traps

PMA

phorbol-12-myristate-13-acetate

PPP

platelet-poor plasma

PRP

platelet-rich plasma

TLR

toll-like receptor

Acknowledgments

T.J. Gould and T.T. Vu performed the experiments. T. J. Gould and P.C. Liaw wrote the paper. L.L. Swystun, T.T. Vu, D.J. Dwivedi, J.I. Weitz, S.H.C. Mai, and P.C. Liaw edited the paper. J.I. Weitz and P.C. Liaw designed the research study. We are extremely grateful to Dr Alison Fox-Robichaud, Dr Deborah Cook, Ellen McDonald, Nicole Zytaruk, and Bronwyn Cash-Barlow for the recruitment of patients with sepsis in Hamilton, Ontario, Canada.

Footnotes

The online-only Data Supplement is available with this article at http://atvb.ahajournals.org/lookup/suppl/doi:10.1161/ATVBAHA.114.304114/-/DC1.

Correspondence to Patricia Liaw, PhD, Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St E, Room C5-107 (5th floor), Hamilton, Ontario L8L 2X2, Canada. E-mail

References

  • 1. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR . Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care.Crit Care Med. 2001; 29:1303–1310.CrossrefMedlineGoogle Scholar
  • 2. Wheeler AP, Bernard GR . Treating patients with severe sepsis.N Engl J Med. 1999; 340:207–214.CrossrefMedlineGoogle Scholar
  • 3. Hotchkiss RS, Karl IE . The pathophysiology and treatment of sepsis.N Engl J Med. 2003; 348:138–150.CrossrefMedlineGoogle Scholar
  • 4. Ulloa L, Brunner M, Ramos L, Deitch EA . Scientific and clinical challenges in sepsis.Curr Pharm Des. 2009; 15:1918–1935.CrossrefMedlineGoogle Scholar
  • 5. Dellinger RP, Levy MM, Carlet JM, et al . Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008.Intensive Care Med. 2008; 34:17–60.CrossrefMedlineGoogle Scholar
  • 6. Yipp BG, Kubes P . NETosis: how vital is it?Blood. 2013; 122:2784–2794.CrossrefMedlineGoogle Scholar
  • 7. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A . Neutrophil extracellular traps kill bacteria.Science. 2004; 303:1532–1535.CrossrefMedlineGoogle Scholar
  • 8. Fuchs TA, Brill A, Wagner DD . Neutrophil extracellular trap (NET) impact on deep vein thrombosis.Arterioscler Thromb Vasc Biol. 2012; 32:1777–1783.LinkGoogle Scholar
  • 9. Swystun LL, Mukherjee S, Liaw PC . Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus.J Thromb Haemost. 2011; 9:2313–2321.CrossrefMedlineGoogle Scholar
  • 10. von Brühl ML, Stark K, Steinhart A, et al . Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo.J Exp Med. 2012; 209:819–835.CrossrefMedlineGoogle Scholar
  • 11. Clark SR, Ma AC, Tavener SA, et al . Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood.Nat Med. 2007; 13:463–469.CrossrefMedlineGoogle Scholar
  • 12. Gupta AK, Joshi MB, Philippova M, Erne P, Hasler P, Hahn S, Resink TJ . Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death.FEBS Lett. 2010; 584:3193–3197.CrossrefMedlineGoogle Scholar
  • 13. Kawasaki H, Iwamuro S . Potential roles of histones in host defense as antimicrobial agents.Infect Disord Drug Targets. 2008; 8:195–205.CrossrefMedlineGoogle Scholar
  • 14. 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. 2009; 15:1318–1321.CrossrefMedlineGoogle Scholar
  • 15. Xu J, Zhang X, Monestier M, Esmon NL, Esmon CT . Extracellular histones are mediators of death through TLR2 and TLR4 in mouse fatal liver injury.J Immunol. 2011; 187:2626–2631.CrossrefMedlineGoogle Scholar
  • 16. Dwivedi DJ, Toltl LJ, Swystun LL, Pogue J, Liaw KL, Weitz JI, Cook DJ, Fox-Robichaud AE, Liaw PC ; the Canadian Critical Care Translational Biology Group. Prognostic utility and characterization of cell-free DNA in patients with severe sepsis.Crit Care. 2012; 16:R151.CrossrefMedlineGoogle Scholar
  • 17. 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. 2013; 123:2981–2993.CrossrefMedlineGoogle Scholar
  • 18. 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. 2010; 207:1853–1862.CrossrefMedlineGoogle Scholar
  • 19. Semeraro F, Ammollo CT, Morrissey JH, Dale GL, Friese P, Esmon NL, Esmon CT . Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4.Blood. 2011; 118:1952–1961.CrossrefMedlineGoogle Scholar
  • 20. Elzagallaai A, Rosé SD, Trifaró JM . Platelet secretion induced by phorbol esters stimulation is mediated through phosphorylation of MARCKS: a MARCKS-derived peptide blocks MARCKS phosphorylation and serotonin release without affecting pleckstrin phosphorylation.Blood. 2000; 95:894–902.CrossrefMedlineGoogle Scholar
  • 21. Lo YM, Rainer TH, Chan LY, Hjelm NM, Cocks RA . Plasma DNA as a prognostic marker in trauma patients.Clin Chem. 2000; 46:319–323.CrossrefMedlineGoogle Scholar
  • 22. Rainer TH, Wong LK, Lam W, Yuen E, Lam NY, Metreweli C, Lo YM . Prognostic use of circulating plasma nucleic acid concentrations in patients with acute stroke.Clin Chem. 2003; 49:562–569.CrossrefMedlineGoogle Scholar
  • 23. Chang CP, Chia RH, Wu TL, Tsao KC, Sun CF, Wu JT . Elevated cell-free serum DNA detected in patients with myocardial infarction.Clin Chim Acta. 2003; 327:95–101.CrossrefMedlineGoogle Scholar
  • 24. Zeerleder S, Zwart B, Wuillemin WA, Aarden LA, Groeneveld AB, Caliezi C, van Nieuwenhuijze AE, van Mierlo GJ, Eerenberg AJ, Lämmle B, Hack CE . Elevated nucleosome levels in systemic inflammation and sepsis.Crit Care Med. 2003; 31:1947–1951.CrossrefMedlineGoogle Scholar
  • 25. Margraf S, Lögters T, Reipen J, Altrichter J, Scholz M, Windolf J . Neutrophil-derived circulating free DNA (cf-DNA/NETs): a potential prognostic marker for posttraumatic development of inflammatory second hit and sepsis.Shock. 2008; 30:352–358.CrossrefMedlineGoogle Scholar
  • 26. 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. 2012; 7:e32366.CrossrefMedlineGoogle Scholar
  • 27. Meng W, Paunel-Görgülü A, Flohé S, Hoffmann A, Witte I, Mackenzie C, Baldus SE, Windolf J, Lögters TT . Depletion of neutrophil extracellular traps in vivo results in hypersusceptibility to polymicrobial sepsis in mice.Crit Care. 2012; 16:R137.CrossrefMedlineGoogle Scholar
  • 28. McDonald B, Urrutia R, Yipp BG, Jenne CN, Kubes P . Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis.Cell Host Microbe. 2012; 12:324–333.CrossrefMedlineGoogle Scholar
  • 29. Coenraad Hemker H, Dieri RA, De Smedt E, Béguin S . Thrombin generation, a function test of the haemostaticthrombotic system.Thromb Haemost. 2006; 96: 553–561.CrossrefMedlineGoogle Scholar
  • 30. Picoli-Quaino SK, Alves BE, Faiotto VB, Montalvao SA, De Souza CA, Annichino-Bizzacchi JM, De Paula EV . Impairment of thrombin generation in the early phases of the host response of sepsis.J Crit Care. 2014; 29:31–36.CrossrefMedlineGoogle Scholar
  • 31. Petros S, Kliem P, Siegemund T, Siegemund R . Thrombin generation in severe sepsis.Thromb Res. 2012; 129:797–800.CrossrefMedlineGoogle Scholar
  • 32. Collins PW, Macchiavello LI, Lewis SJ, Macartney NJ, Saayman AG, Luddington R, Baglin T, Findlay GP . Global tests of haemostasis in critically ill patients with severe sepsis syndrome compared to controls.Br J Haematol. 2006; 135:220–227.CrossrefMedlineGoogle Scholar
  • 33. Massion PB, Peters P, Ledoux D, Zimermann V, Canivet JL, Massion PP, Damas P, Gothot A . Persistent hypocoagulability in patients with septic shock predicts greater hospital mortality: impact of impaired thrombin generation.Intensive Care Med. 2012; 38:1326–1335.CrossrefMedlineGoogle Scholar
  • 34. Seo JW, Kim HK, Kim JE, Park S, Cho HI . Prognostic values of the factor Xa-activated clotting time and endogenous thrombin potential in patients suspected of having disseminated intravascular coagulation.Thromb Res. 2009; 123:565–572.CrossrefMedlineGoogle Scholar
  • 35. Hodson ME, Shah PL . DNase trials in cystic fibrosis.Eur Respir J. 1995; 8:1786–1791.CrossrefMedlineGoogle Scholar
  • 36. Macanovic M, Sinicropi D, Shak S, Baughman S, Thiru S, Lachmann PJ . The treatment of systemic lupus erythematosus (SLE) in NZB/W F1 hybrid mice; studies with recombinant murine DNase and with dexamethasone.Clin Exp Immunol. 1996; 106:243–252.CrossrefMedlineGoogle Scholar
  • 37. Higuchi DA, Wun TC, Likert KM, Broze GJ . The effect of leukocyte elastase on tissue factor pathway inhibitor.Blood. 1992; 79:1712–1719.CrossrefMedlineGoogle Scholar
  • 38. Massberg S, Grahl L, von Bruehl ML, et al . Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases.Nat Med. 2010; 16:887–896.CrossrefMedlineGoogle Scholar
  • 39. Kleine TJ, Gladfelter A, Lewis PN, Lewis SA . Histone-induced damage of a mammalian epithelium: the conductive effect.Am J Physiol. 1995; 268(5 Pt 1):C1114–C1125.CrossrefMedlineGoogle Scholar
  • 40. Peters E, van Elsas A, Heemskerk S, Jonk L, van der Hoeven J, Arend J, Masereeuw R, Pickkers P . Alkaline phosphatase as a treatment of sepsis-associated acute kidney injury.J Pharmacol Exp Ther. 2012; 344:2–7.CrossrefMedlineGoogle Scholar
  • 41. Heemskerk S, Masereeuw R, Moesker O, Bouw MP, van der Hoeven JG, Peters WH, Russel FG, Pickkers P ; APSEP Study Group. Alkaline phosphatase treatment improves renal function in severe sepsis or septic shock patients.Crit Care Med. 2009; 37:417–423, e1.CrossrefMedlineGoogle Scholar
  • 42. Pickkers P, Heemskerk S, Schouten J, Laterre PF, Vincent JL, Beishuizen A, Jorens PG, Spapen H, Bulitta M, Peters WH, van der Hoeven JG . Alkaline phosphatase for treatment of sepsis-induced acute kidney injury: a prospective randomized double-blind placebo-controlled trial.Crit Care. 2012; 16:R14.CrossrefMedlineGoogle Scholar

Significance

These studies examine the procoagulant activities of cell-free DNA and histones in the context of intact neutrophil extracellular traps (NETs). We demonstrated that intact NETs promote thrombin generation in platelet-poor plasma and that (1) thrombin generation is triggered via the intrinsic pathway, and (2) thrombin generation in platelet-poor plasma is attenuated with DNase but not RNase. Thrombin generation in PRP containing intact NETs was further enhanced with DNase addition, an effect that was abolished with anti–toll-like receptor antibodies. This suggests that degradation of cell-free DNA in the NET scaffold increases histone-mediated, platelet-dependent thrombin generation. Our studies also implicate a role for cell-free DNA–mediated activation of the intrinsic pathway of coagulation in the pathogenesis of sepsis. We observed a direct correlation between plasma CFDNA levels and indices of thrombin generation in patients with sepsis. Our findings support the concept that NET components may be important therapeutic targets for the treatment of sepsis.