In recent years, the traditional view of the hemostatic system as being regulated by a coagulation factor cascade coupled with platelet activation has been increasingly challenged by new evidence that activation of the immune system strongly influences blood coagulation and pathological thrombus formation. Leukocytes can be induced to express tissue factor and release proinflammatory and procoagulant molecules such as granular enzymes, cytokines, and damage-associated molecular patterns. These mediators can influence all aspects of thrombus formation, including platelet activation and adhesion, and activation of the intrinsic and extrinsic coagulation pathways. Leukocyte-released procoagulant mediators increase systemic thrombogenicity, and leukocytes are actively recruited to the site of thrombus formation through interactions with platelets and endothelial cell adhesion molecules. Additionally, phagocytic leukocytes are involved in fibrinolysis and thrombus resolution, and can regulate clearance of platelets and coagulation factors. Dysregulated activation of leukocyte innate immune functions thus plays a role in pathological thrombus formation. Modulation of the interactions between leukocytes or leukocyte-derived procoagulant materials and the traditional hemostatic system is an attractive target for the development of novel antithrombotic strategies.

Hemostatic thrombus formation is conventionally thought to involve a coagulation factor cascade coupled with platelet activation. Pathological thrombosis, as described in the 1850s by the German pathologist Rudolph Virchow, is influenced by aberrant activation of coagulation, disruption of the vessel wall, and stasis.1  However, in recent years this model has undergone a significant paradigm shift due to accumulating evidence of an intrinsic link between the coagulation and innate inflammatory systems. The term immunothrombosis, coined in 2013 by Engelmann and Massberg,2  formalized this concept and described a process by which the activation of coagulation assists the function of the innate immune system, and the converse, whereby components of the immune system contribute to thrombosis. Dysregulated activation of the immune system can thus contribute to the genesis of pathological macro- and microvascular thrombosis.

The contribution of leukocytes to coagulation is a subject of both longstanding interest as well as current intensive study. With the development of intravital imaging techniques, animal models that closely mimic the pathogenesis of thrombosis in humans, and selective antagonists of leukocyte-regulated procoagulant pathways, the role that leukocytes play in regulating thrombosis is being unveiled. Leukocytes, namely monocytes, macrophages, and neutrophils, express and release coagulation and fibrinolytic factors, and interact with the hemostatic system through innate immune functions. Leukocytes produce cytokines that modulate the expression of procoagulant and adhesive molecules on vascular endothelial cells. Antimicrobial agents released during leukocyte degranulation and extracellular trap formation directly activate platelets and the coagulation cascade. Additionally, leukocyte chemotaxis and phagocytic functions regulate thrombus resolution. In this review, the influence of leukocytes on blood coagulation and platelet activation will be described, and evidence assessing the contribution of leukocytes to venous, arterial, and microvascular thrombosis will be considered.

Under normal physiological circumstances, quiescent leukocytes promote the maintenance of blood fluidity. For example, circulating monocytes express the anticoagulant factors endothelial protein C receptor (EPCR),3  thrombomodulin (TM),4  and tissue factor pathway inhibitor (TFPI).5  However, under proinflammatory or apoptotic conditions, leukocytes can rapidly undergo a phenotypic transformation, synthesizing and secreting procoagulant factors or agents that activate coagulation (Figure 1). Additionally, the leukocyte cell surface can provide a site for coagulation factor assembly and activation.6,7 

Figure 1

Leukocyte-released enzymes and DAMPs interact with components of the coagulation cascade. CF-DNA, histones, MMP, cathepsin G, and/or elastase modulate the traditional coagulation cascade by facilitating the activation of zymogen coagulation and fibrinolytic factors, and inhibiting the activity of endogenous anticoagulants. TF expressed by circulating leukocytes can activate coagulation through the extrinsic pathway, whereas monocyte-derived uPA can modulate fibrinolysis. CF-DNA, cell-free DNA; MMP, matrix metalloproteinase; PAI, plasminogen activator inhibitor; uPA, urokinase-type plasminogen activator.

Figure 1

Leukocyte-released enzymes and DAMPs interact with components of the coagulation cascade. CF-DNA, histones, MMP, cathepsin G, and/or elastase modulate the traditional coagulation cascade by facilitating the activation of zymogen coagulation and fibrinolytic factors, and inhibiting the activity of endogenous anticoagulants. TF expressed by circulating leukocytes can activate coagulation through the extrinsic pathway, whereas monocyte-derived uPA can modulate fibrinolysis. CF-DNA, cell-free DNA; MMP, matrix metalloproteinase; PAI, plasminogen activator inhibitor; uPA, urokinase-type plasminogen activator.

Close modal

Tissue factor (TF)

Monocytes are the largest intravascular source of TF.8,9  Although low levels of TF antigen are detected on quiescent monocytes, exposure to agents that promote inflammation, and/or apoptosis, including high mobility group box-1 (HMGB-1),10  chemotherapy,11  lipopolysaccharide,12  hypoxia,13  and anti-HIT antibodies14  increase monocyte TF activity and/or TF-mediated thrombin generation. Monocyte TF activity is regulated by processes that increase TF expression, induce TF decryption,15  and modulate the balance between TF and TFPI.16  Activated monocytes also shed microparticles from P-selectin glycoprotein ligand 1 (PSGL-1)–rich membrane microdomains17  that carry TF, phosphatidylserine, and other regulators of coagulation.18  Expression of TF by other leukocyte subtypes is more controversial. Although quiescent neutrophils likely do not express TF antigen,19  TF can be expressed in smaller quantities by neutrophils stimulated ex vivo20,21  and in animal models.22,23  This may in part be the result of neutrophil acquisition of TF from monocytes,24  potentially through a process involving microparticle-mediated transfer. Similarly, conflicting reports describe the expression of TF by eosinophils, but may partially explain the increased risk for thrombosis in eosinophilia.25-27 

Granular enzymes

Neutrophils, and to a lesser extent monocytes, and basophils release matrix metalloproteinases and serine proteases such as cathepsin G and elastase, from cytoplasmic granules in response to stimulation.28,29  These enzymes promote coagulation activation through numerous mechanisms (Figure 1), including directly activating cofactors factor V (FV),30  FVIII,31  and zymogen FX.32  They can also degrade anticoagulant factors such as antithrombin,33  heparin cofactor II,34  and/or TFPI.35-37 

Nuclear damage-associated molecular patterns (DAMPs)

DAMPs, including DNA, HMGB1, and histones are released from the nuclei of activated or apoptotic leukocytes and promote the activation of coagulation. The release of neutrophil chromatin as neutrophil extracellular traps (NETs) can be triggered by exposure to microorganisms, activated platelets, inflammatory cytokines, and HMGB1.38-41  As well, NETs have also been reported to be released by monocytes/macrophages42  and mast cells,43  whereas basophils44  and eosinophils45  release extracellular traps comprised of granular enzymes and mitochondrial DNA. CF-DNA may also be released from apoptotic or necrotic cells in the circulation or vessel wall.46 

Intact NETs act as a scaffold that concentrates procoagulant effectors including platelets, red blood cells, von Willebrand factor (VWF), TF, protein disulfide isomerase, HMGB1, cathepsin G, elastase, fibrin(ogen), and fibronectin.22,47,48  The influence of NET components on coagulation activation has been independently evaluated (Figure 1). Extracellular DNA triggers contact pathway activation through FXI and FXII.49,50  Histone H4 binds to prothrombin and generates thrombin by auto-activation.51  DAMPs can also inhibit anticoagulant pathways by protecting thrombin from antithrombin-mediated inactivation,52,53  and by impairing protein C activation by thrombin-TM.10,54 

Leukocyte-released antimicrobial enzymes, cytokines, and DAMPs, can modulate the anticoagulant activity of endothelial cells (Figure 2). Cytokines such as tumor necrosis factor-α and interleukin-1β can downregulate expression of EPCR and TM through decreased messenger RNA synthesis,55  and increased EPCR shedding.56  Histones are cytotoxic to endothelial cells,57  and increase surface phosphatidyserine exposure on eryothrocytes.58  Histones,59  cytokines,60  HMGB1,10  and granular enzymes61  can increase in vitro endothelial cell TF activity. Histones and cytokines can also stimulate the exocytosis of endothelial Weibel-Palade bodies, inducing the release of VWF and/or P-selectin.62,63  Activated basophils also release histamine,29  a potent secretagogue for VWF.64  Additionally, both cytokines and neutrophil-generated oxidants such as HOCl impair cleavage of VWF by the protease ADAMTS13, potentially increasing the proportion of circulating ultralarge VWF multimers with enhanced platelet-binding abilities.63,65 

Figure 2

Leukocyte-released enzymes, DAMPs, and cytokines regulate the hemostatic activity of endothelial cells and platelets. Mediators released by activated leukocytes can influence the hemostatic activity of the endothelium and platelet activation. WPB, Weibel-Palade body.

Figure 2

Leukocyte-released enzymes, DAMPs, and cytokines regulate the hemostatic activity of endothelial cells and platelets. Mediators released by activated leukocytes can influence the hemostatic activity of the endothelium and platelet activation. WPB, Weibel-Palade body.

Close modal

Although activated platelets can stimulate NETosis,41  leukocytes interact with activated platelets to form heterotypic leukocyte-platelet aggregates (Figure 3B). Exposure to proinflammatory and procoagulant stimuli, as well as high shear stress have been shown to promote the formation of these complexes.66-69  Heterotypic aggregates form when activated, and degranulated platelets expose P-selectin on their surface that binds to leukocyte surface PSGL-1.70  Signaling through PSGL-1 rapidly upregulates leukocyte expression of αMβ271  that binds platelet GPIbα72  or GPIIbIIIa via a fibrinogen intermediate (Figure 3B).73  The complexes are further stabilized by multiple receptor-ligand interactions including CD40-CD40 ligand,74  extracellular matrix metalloproteinase inducer-glycoprotein VI (GPVI),75  lymphocyte function-associated antigen 1-ICAM-2,76  and junctional adhesion molecule-C-αMβ2.77  PSGL-1 engagement can also activate cooperative signaling through NF-κB to induce the production of proinflammatory cytokines.78  Although conflicting reports exist, evidence suggests that platelet P-selectin interactions with leukocyte PSGL-1 may also activate TF on monocytes and/or neutrophils.20,78,79  Increased levels of leukocyte-platelet aggregates are frequently associated with thromboinflammatory disorders, and can be used as stable markers of underlying hypercoagulability.

Figure 3

Recruitment of leukocytes to the growing thrombus. Leukocyte recruitment to the thrombus is influenced by the condition of the endothelium and the shear rate in the vessel. (A) Under low shear with intact endothelium, leukocytes are able to bind adhesive molecules on the endothelial surface such as VWF, selectin family members, and cell adhesion molecules to leukocyte-expressed PSGL-1 or integrin receptors. (B) In arterial thrombosis, leukocyte recruitment to the thrombus is influenced by platelet binding, where glycoprotein receptors/β1 and β3 integrins on activated platelets bind to subendothelial adhesion molecules such as VWF, collagen, laminin, and fibronectin. Leukocytes bind to adhered platelets in a PSGL-1/P-selectin–dependent manner and are further stabilized by additional ligand-receptor interactions. EMMPRIN, extracellular matrix metalloproteinase inducer; JAM-3, junctional adhesion molecule; LFA-1, lymphocyte function-associated antigen 1.

Figure 3

Recruitment of leukocytes to the growing thrombus. Leukocyte recruitment to the thrombus is influenced by the condition of the endothelium and the shear rate in the vessel. (A) Under low shear with intact endothelium, leukocytes are able to bind adhesive molecules on the endothelial surface such as VWF, selectin family members, and cell adhesion molecules to leukocyte-expressed PSGL-1 or integrin receptors. (B) In arterial thrombosis, leukocyte recruitment to the thrombus is influenced by platelet binding, where glycoprotein receptors/β1 and β3 integrins on activated platelets bind to subendothelial adhesion molecules such as VWF, collagen, laminin, and fibronectin. Leukocytes bind to adhered platelets in a PSGL-1/P-selectin–dependent manner and are further stabilized by additional ligand-receptor interactions. EMMPRIN, extracellular matrix metalloproteinase inducer; JAM-3, junctional adhesion molecule; LFA-1, lymphocyte function-associated antigen 1.

Close modal

Activated leukocytes can also induce platelet activation and aggregation by releasing potent platelet activators including elastase,80  cathepsin G,81  and platelet activating factor (Figure 2).82  The influence of NETs on platelet activation has been characterized using several in vitro and in vivo models. NETs can bind to both platelets and VWF under shear,47  and stimulation of endothelial cells by histones increases platelet capture by VWF in a flow chamber system.62  This interaction is likely mediated by both increased Weibel-Palade body exocytosis and platelet activation, as both intact NETs50  and extracellular histones induce platelet activation via toll-like receptor 2 (TLR2) and TLR4.83,84  Platelet activation by histones promote the formation of leukocyte-platelet aggregates and induces the release of platelet VWF, P-selectin, and polyphosphate, which promotes platelet-dependent thrombin generation.69,84  In vivo, infusions of extracellular histones promote the formation of platelet-rich microthrombi with concomitant thrombocytopenia.83 

Clinical studies have described evidence of activated leukocytes associated with venous, arterial, and microvascular thrombosis. Elevated levels of circulating markers of NETs and neutrophil activation,85,86  as well as increased monocyte TF,87,88  have been observed in patients with DVTs compared with control subjects. In addition to releasing disseminated procoagulant factors into the blood, it is increasingly recognized that leukocytes also assemble at the site of vascular injury, and are actively incorporated into forming thrombi. Studies evaluating the composition of venous thrombi from humans demonstrated the presence of TF-expressing leukocytes and NETs.89,90  These processes increase the localized concentration of leukocyte-derived procoagulant activity and potentially forms the nidus upon which the thrombus develops.

Experimental animal models have also shown the association of leukocyte recruitment with the induction of venous thrombosis. In the detailed characterization of a mouse inferior vena cava (IVC) stenosis model, the recruitment of leukocytes to the site of venous thrombosis occurred overlying the intact but activated endothelium within an hour of vessel flow restriction.22  Within 6 hours, leukocytes overlay the endothelial surface, with neutrophils and monocytes comprising 70% and 30% of leukocytes within the thrombus, respectively. Leukocyte recruitment may also be facilitated by the release of cytokines and chemokines by activated platelets,91  which bind directly to the endothelium, or form heterotypic aggregates within the venous thrombus.22,92  Leukocyte rolling is mediated by the upregulation of selectins on the endothelial surface that binds leukocyte PSGL-1, and genetic deletion of P- and E-selectins reduces leukocyte accumulation and venous thrombus size (Figure 3A).22,93  Firm attachment of leukocytes can involve endothelial VWF, VCAM, and ICAM binding leukocyte PSGL-1 and integrins.22,94-96 

Assessment of the contribution of leukocytes to the genesis of venous thrombosis is dependent on the animal model used. Both neutrophil and monocyte depletion inhibit thrombosis in murine IVC stenosis22  and ferric chloride models,97  respectively. With respect to leukocyte TF, infusion of activated TF-expressing monocytes resulted in systemic venous and arterial thrombosis in rabbits,98  and IVC ligation models have demonstrated the presence of TF-expressing leukocytes within venous thrombi in rabbits99  and rats.100  In the venous stenosis IVC ligation model, selective depletion of hematopoietic TF significantly attenuates thrombus formation22 ; however, in contrast, DVT formation in response to complete IVC stasis does not involve hematopoietic TF.101 

Although the majority of leukocyte-released TF associated with venous thrombi is likely derived from monocytes, neutrophils have also been variably implicated in the formation of DVT. For example, NETs are associated with venous thrombi derived from murine and baboon models.102,103  Infusions of extracellular histones promote DVT development in murine IVC stenosis models, whereas the administration of DNAse1, or FXII deficiency attenuates DVT formation.22,103  In contrast, a model of spontaneous venous thrombosis using small interfering RNA knockdown of protein C and antithrombin, showed that although neutrophils were associated with venous thrombi, neutrophil depletion did not diminish thrombus formation.104  Interestingly, protein arginine deiminase 4 (PAD4) and neutrophil elastase, which regulate chromatin decondensation and NETosis, have contradictory influence on DVT induction. Although PAD4 deficiency was associated with impaired thrombus formation in the IVC stenosis model,105  an electrolytic injury model reported no influence of PAD4 deficiency on DVT.106  Additionally, neutrophil elastase deficiency has been shown to impair NET formation in response to microbial infection,107,108  however, a sterile IVC stenosis model demonstrated elastase-deficient mice generate NETs and have normal venous thrombi.109  Although overall these studies appear to support a role for leukocytes in the induction of thrombosis in response to venous stenosis, they also highlight how the variability of the models employed, including the extent of endothelial injury and underlying activation status of circulating leukocytes, can influence the induction of venous thrombosis.

Leukocytes can participate in atherothrombosis by generating procoagulant material within the atherosclerotic plaque and contributing to the formation of arterial thrombi overlying the site of vessel rupture. Localized exposure of vessel wall procoagulant material, including macrophage/foam cells that express high levels of TF110  and shed TF-positive microparticles,111  is the precipitating event in atherothrombosis. Arterial thrombosis occurs under conditions of high shear, whereby circulating platelets bind subendothelial ligands, such as collagen and VWF that facilitate the generation of a platelet-rich thrombus. Activated platelets release cytokines that can modulate leukocyte activation,41,112  resulting in the formation of heterotypic leukocyte-platelet aggregates that serve to localize activated leukocytes to the arterial thrombus (Figure 3B). Plasma levels of TF-expressing monocytes and microparticles, leukocyte-platelet aggregates, and NET markers are elevated in conditions that predispose to cardiovascular disease.113,114  Histologic analysis of coronary artery and catheter-associated arterial thrombi demonstrated the presence of monocytes, neutrophils, and eosinophils within the thrombi.115,116 

Although murine models do not typically display spontaneous atherothrombosis, intravital models of arterial thrombosis address thrombus formation in macrovascular beds in the context of activated or damaged endothelium. Vessel wall TF is unequivocally involved in thrombus initiation in arterial thrombosis,117  however, the role of circulating leukocyte or leukocyte-derived TF regulating arterial thrombus formation is less clear. The inhibition of leukocyte accumulation using an anti–P-selectin antibody in a baboon model of arterial thrombus significantly attenuated fibrin formation and decreased thrombus stability.118  Another study utilizing reciprocal bone marrow transplants between normal and low TF-expressing mice showed no decrease in Rose Bengal-induced carotid artery thrombus formation in healthy mice in the absence of hematopoietic TF.101  However, infusion of microparticles prepared from human monocytes increased fibrin formation in a carotid ligation injury in a TF-dependent manner.119  Because monocyte and microparticle TF activity is generally increased for individuals predisposed to arterial thrombosis, the direct influence of hematopoietic TF on arterial thrombus formation in this context has yet to be clearly elucidated.

The role of neutrophils in the propagation of arterial thrombosis has been characterized in murine carotid artery ferric chloride and ligation models.35  In these studies, elastase/cathepsin G-deficient mice had reduced arterial thrombus formation related to serine-protease degradation of TFPI. Moreover, an anti-H2A-H2B-DNA neutralizing antibody impaired thrombus formation and decreased thrombus stability in normal but not elastase/cathepsin G-deficient mice. This effect may be related to the reduction in co-assembly of elastase, cathepsin G, and TFPI with extracellular nucleosomes,35  although the influence of elastase deficiency on NET formation was not directly evaluated in this model. In animal models, NETs are associated with the lumen overlying the atherosclerotic plaque,120  and are found elaborated with TF within the arterial thrombus, suggesting that NETs may help localize leukocyte-derived TF within the arterial thrombus.121  Interestingly, the administration of DNAse does not attenuate arterial thrombus formation in healthy mice,122  although both DNAse and/or PAD4 inhibition impair arterial thrombosis in murine models of lupus and atherosclerosis, where animals are predisposed to NET formation.123,124 

Microvascular thrombosis involves the development of thrombi in the venules, arterioles, and capillaries. Individuals with thrombotic microangiopathies arising from non-infectious etiologies display evidence of elevated NET markers125  and impaired DNAse function126  that may contribute to the acute phase of these disorders. In healthy animals, microvascular thrombosis is evaluated most frequently using chemical or laser-induced endothelial injury models of the cremaster and mesenteric arterioles. In these models, it has been demonstrated that TF delivery to the thrombus is mediated through accumulation of both leukocytes23  as well as leukocyte-derived microparticles.127,128  Adhesion of neutrophils to the activated endothelium occurs immediately, and is mediated through lymphocyte function-associated antigen 1 binding to endothelial ICAM-1, whereas monocyte recruitment occurs 3 to 5 minutes post-injury.23  Depletion of neutrophils or impaired neutrophil-endothelial cell interactions diminished TF accumulation and thrombus formation, suggesting that TF-positive neutrophils form a focus for thrombus development.23  Concurrently, TF-expressing microparticles from both vessel wall and leukocyte origin accumulate at the injury in a PSGL-1/P-selectin–dependent manner.128,129  Monocyte and microparticle-associated TF may facilitate thrombus propagation, because in the absence of hematopoietic TF, fibrin deposition throughout the thrombus is diminished.128 

In humans, microvascular thrombosis is most frequently associated with DIC caused by endotoxemia, sepsis, or trauma. Evidence suggests that dysregulated activation of leukocytes is associated with sepsis and/or DIC in humans and animal models. Endotoxin and/or microbes can stimulate the expression of TF on monocytes,130,131  formation of leukocyte-derived microparticles,132  neutrophil degranulation, and NETosis41  ex vivo. Patients with sepsis and/or evidence of DIC have evidence of elevated monocyte TF,133  TF-expressing microparticles,134  HMGB1,135  NET markers,136  and leukocyte-platelet aggregates.137 

In mice, hematopoietic cell TF contributes to the coagulopathy observed models of endotoxemia.138  Infusions of extracellular histones in mice mimic the pathophysiology of DIC by inducing thrombocytopenia, the formation of platelet-rich microthrombi,83  and microvascular thrombosis, with concomitant bleeding.57  Additionally, elastase/cathepsin G-deficient mice demonstrated decreased fibrin deposition in response to Escherichia coli infection, and treatment with an anti-H2A-H2B-DNA antibody diminished fibrin production and microvascular occlusions.35  Similarly, infusion of exogenous HMGB1 can potentiate thrombosis and hemorrhage in kidney and lung microvasculature in a thrombin-induced model of DIC.10 

In addition to influencing thrombus induction, leukocytes regulate thrombus persistence and levels of activated or acute phase coagulation factors in the circulation. This includes modulation of fibrinolysis through expression and activation of fibrinolytic mediators. Additionally, leukocytes regulate thrombus resolution and coagulation factor clearance through phagocytosis.

Fibrinolysis

Both monocytes and neutrophils can modulate activity of the fibrinoytic pathway and susceptibility of formed fibrin to fibrinolysis (Figure 1). In vitro fibrinolysis is accelerated in the presence of isolated quiescent neutrophils139  and monocytes,140  and monocyte-derived microparticles141  through several characterized mechanisms. Leukocytes express uPA and its receptor uPAR,142  and hematopoietic uPA deficiency is associated with attenuated thrombus resolution in vivo.143  Leukocytes also express receptors for plasminogen, including enolase, Annexin II, and histone H2B, which localize plasminogen to the leukocyte surface, thereby enhancing activation by tissue-type plasminogen activator (tPA) and/or uPA.144  Additionally, elastase has been shown to inactivate plasminogen activator inhibitor145  and activate plasmin in the absence of tPA/uPA.146 

However, under pathological circumstances, activated leukocytes may attenuate endogenous fibrinolytic mechanisms. Lipopolysaccharide-stimulated monocytes inhibit fibrinolysis by increasing activation of thrombin activatable fibrinolysis inhibitor in a TF-dependent manner.147  NETosis may also influence fibrinolysis and thrombus stability, as the addition of CF-DNA and histones to clotting plasma results in the formation of thicker fibers with greater mechanical stability.148  In patients with sepsis and elevated CF-DNA levels, clot lysis times are attenuated compared with controls149 ; this effect can be replicated by the addition of histone-DNA complex148  or in the presence of NETing neutrophils.53  The presence of CF-DNA within the clot has been shown to impair plasminogen activation by tPA,53  and the binding of plasmin to fibrin.150 

Phagocytosis

In addition to modulation of fibrinolysis, phagocytic leukocytes play a vital role in regulating the persistence of active coagulation factors and formed thrombi within the vasculature. Plasma levels of coagulation factors strongly influence the propensity for pathological thrombus formation, and regulation of plasma levels of the coagulation factors involves a dynamic balance between biosynthesis, secretion, and clearance. Monocyte and/or macrophages express scavenger receptors, including low-density lipoprotein receptor family (eg, low density lipoprotein receptor-related protein 1), sialic acid-binding immunoglobulin-type lectin family members , and αMβ2 integrin, which regulate the endocytosis of coagulation factors such as VWF and FVIII,151,152  activated platelets,153  fibrin(ogen),154  and/or NETs.108,155  For example, the depletion of macrophages in vivo is associated with elevated plasma levels of VWF-FVIII,151  a risk-factor for venous and arterial thrombosis. Neutrophils can also contribute to the clearance of activated platelets through interactions mediated by platelet surface P-selectin binding to neutrophil-expressed PSGL-1. This interaction is stabilized by neutrophil-expressed β2 integrins, and requires phosphatidylserine exposure on the platelet surface to mediate endocytosis of the active platelet.156 

Leukocytes also mediate venous thrombus resolution, a process that in addition to fibrinolysis and phagocytosis involves angiogenesis, fibrosis, and vessel wall remodeling.157,158  Circulating leukocytes are recruited to the thrombus by release of proinflammatory cytokines and chemokines, upregulated adhesion molecules on the endothelium,159  the binding of plasminogen to plasminogen receptors, and the formation of fibrin.160-162  Leukocyte infiltration is temporally regulated with neutrophils predominating at early stages, and monocytes and macrophages predominating at later stages.159  In animal models involving neutrophil depletion, IVC ligation in rats was associated with delayed thrombus resolution,157  although this effect is not observed in mice. However, impaired recruitment of monocytes to the resolving thrombus is associated with increased thrombus size, and decreased neovascularization in murine IVC stasis models.158,163 

Although the role of neutrophils in thrombus resolution is not fully characterized, they may contribute to this process by phagocytosing apoptotic cells and by-products of thrombolysis. Monocytes have been shown to regulate thrombus resolution by influencing fibrinolysis, producing growth factors, matrix metalloproteinases,158  and uPA,164  which in addition to activating plasminogen mediates cell migration and tissue remodeling.143  Importantly, there may be heterogeneous roles for monocyte subtypes in the process of thrombus resolution. Ly6C+ monocytes, considered to be proinflammatory, are recruited early to damaged tissue, and have been hypothesized to play a role in phagocytosing apoptotic cells and debris associated with the thrombus. Conversely, Ly6C resident monocytes patrol the vasculature, are recruited later to sites of vascular damage, and have been hypothesized to contribute to tissue repair.165,166 

Although leukocyte activation can modify blood coagulation, it is well recognized that active coagulation factors and platelets can also regulate the proinflammatory activity of leukocytes (Figure 4). In the context of thrombosis, this relationship of reciprocal activation can serve to recruit leukocytes to the forming thrombus. Both monocytes and macrophages express protease activated receptor-1 (PAR-1), a G-protein coupled receptor that is activated by coagulation factor proteases.167  Thrombin can induce chemotaxis of both neutrophils168  and monocytes169  via PAR-dependent and -independent mechanisms.170  Conversely, APC inhibits leukocyte chemotaxis through PAR cleavage, and interactions with β1 and β3 integrins.171,172  Active coagulation factors can also mediate the release of procoagulant materials and inflammatory agents by leukocytes. Thrombin can regulate the production of proinflammatory cytokines including interleukin-6 and tumor necrosis factor-α by monocytes through PAR signaling.173  Fibrin and fibrin degradation products can stimulate the release of proinflammatory cytokines from monocytes and macrophages by signaling through TLR4.160,161  Soluble fibrinogen is a potent inducer of neutrophil degranulation via interactions with αMβ2 integrin, and can increase phagocytic activity while delaying neutrophil apoptosis.162  Additionally, activated platelets can interact with neutrophils to induce degranulation156  and NET formation.41  Thus, positive feedback between dysregulated procoagulant and proinflammatory pathways within the developing thrombus may enhance the procoagulant phenotype of thrombus-associated leukocytes and exacerbate the development of pathological thrombosis.

Figure 4

Coagulation factors can activate leukocytes. (A) Thrombin and APC cleave PAR-1 expressed on monocytes, and regulate monocyte proinflammatory and procoagulant properties. (B) Coagulation factors and activated platelets interact with neutrophils to regulate neutrophil degranulation, NET release, and phagocytic and chemotactic activities. APC, activated protein C; PAMP, pathogen-associated molecular pattern.

Figure 4

Coagulation factors can activate leukocytes. (A) Thrombin and APC cleave PAR-1 expressed on monocytes, and regulate monocyte proinflammatory and procoagulant properties. (B) Coagulation factors and activated platelets interact with neutrophils to regulate neutrophil degranulation, NET release, and phagocytic and chemotactic activities. APC, activated protein C; PAMP, pathogen-associated molecular pattern.

Close modal

Current strategies for the clinical management of thrombosis involve the use of prophylactic or on-demand anticoagulant therapies, which are associated with an increased risk for bleeding. Recognition of the contribution of leukocytes to the generation of pathological thrombosis, coupled with limited evidence that leukocytes participate in physiological hemostasis, has resulted in the recent development of rational strategies that specifically target leukocyte-mediated prothrombotic pathways in clinical and preclinical studies. Anti-inflammatory agents such as roflumilast (a phosphodiesterase-4 inhibitor) impair the recruitment of leukocytes to the site of thrombus formation,174  and are associated with a decreased risk for major cardiovascular events in chronic obstructive pulmonary disease patients.175  Additionally, statins, which have pleiotropic anticoagulant effects, including reduced expression of TF by monocytes and attenuated release of TF-expressing monocyte-derived microparticles in animal models of hypercholesterolemia,113  are associated with a decreased incidence of arterial and venous thrombosis in clinical studies.176,177 

Preclinical studies that targeted leukocyte recruitment to the thrombus using monoclonal antibodies to P-selectin murine and primate models demonstrated reduced inflammation and/or venous thrombus formation.178,179  Other strategies have involved inhibiting the procoagulant agents released by activated leukocytes. Treatment with the PAD inhibitor Cl-amidine is capable of blocking NET release, and attenuates thrombus formation and reduces atherosclerotic lesion areas in murine models.123,124  Therapies that decrease the procoagulant activity of histones, including APC,57  recombinant soluble TM,180  non-anticoagulant heparins,181  and neutralizing antibodies,57  protect mice from thrombosis and/or histone-mediated death in animal models of acute inflammation. Dismantling NETs with DNAse is protective from flow-restricted venous thrombosis,103  and arterial thrombosis induced by photochemical injury in a murine model of chronic inflammation.123  Additionally, the utility of DNAse in combination with tPA for thrombolysis has demonstrated efficacy in ex vivo models.102  Nucleic acid-binding polymers, which inhibit nucleic acid- and polyphosphate-induced activation of the intrinsic pathway of coagulation have also been shown to prevent thrombosis in mice without increasing the risk of bleeding.182  Finally, targeting contact pathway activation, such as with monoclonal antibodies to FXII, can reduce thrombus formation in primate models183  and may mitigate some of the procoagulant effects associated with high levels of extracellular DNA.

Leukocytes are a dynamic and itinerant component of the innate immune system that mediate a rapid response to procoagulant stimuli. Localized intravascular coagulation involving activated leukocytes likely evolved as an adaptive mechanism to promote the resolution of infection when trauma and epidemics accounted for the majority of human deaths. Improved sanitation, nutrition, and treatment of infection promoted epidemiological transition, associated with an increased life expectancy, and the development of chronic inflammatory diseases such as atherosclerosis. Thus, the activation of leukocyte procoagulant activity in response to sterile inflammation may be maladaptive, and links the coincidence of micro- and macrovascular thrombosis with inflammatory pathologies.

There remain controversies in the literature that pertain to the heterarchical role neutrophils and monocytes play in thrombus formation that may be related to the model of thrombosis used, as well as the pathobiological context in which the thrombus develops. The continued development of physiologically relevant in vivo models of thrombosis, and careful assessment of the contribution that leukocytes make to thrombo-inflammatory conditions will provide novel insights into the mechanistic basis of thrombosis. These discoveries may be translated to the clinic through improved identification of individuals at-risk for thrombosis. They may also result in the discovery of novel targets for the development of prophylactic or on-demand anticoagulant treatment, and generate strategies for accelerating thrombus resolution. Although modulation of the axis between inflammation and coagulation must take into consideration the potentially beneficial role that leukocyte activities may have in regulating disease outcome and promotion of thrombus resolution, the development of novel inhibitors of leukocyte-associated procoagulant activity may ultimately prove effective at reducing the burden of thrombosis worldwide.

The authors thank T. Gould and L. Pepler for many helpful discussions.

L.L.S. is supported by a postdoctoral fellowship from the Canadian Institutes for Health Research, and P.C.L. is supported by a grant from the Canadian Institutes for Health Research (MOP-136878).

Contribution: L.L.S. created the figures and wrote the manuscript, and P.C.L. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Patricia C. Liaw, Thrombosis and Atherosclerosis Research Institute, McMaster University, 237 Barton St East, Room C5-107, Hamilton, ON L8L 2X2, Canada; e-mail: patricia.liaw@taari.ca.

1
Kumar
 
DR
Hanlin
 
E
Glurich
 
I
Mazza
 
JJ
Yale
 
SH
Virchow’s contribution to the understanding of thrombosis and cellular biology.
Clin Med Res
2010
, vol. 
8
 
3-4
(pg. 
168
-
172
)
2
Engelmann
 
B
Massberg
 
S
Thrombosis as an intravascular effector of innate immunity.
Nat Rev Immunol
2013
, vol. 
13
 
1
(pg. 
34
-
45
)
3
Galligan
 
L
Livingstone
 
W
Volkov
 
Y
, et al. 
Characterization of protein C receptor expression in monocytes.
Br J Haematol
2001
, vol. 
115
 
2
(pg. 
408
-
414
)
4
McCachren
 
SS
Diggs
 
J
Weinberg
 
JB
Dittman
 
WA
Thrombomodulin expression by human blood monocytes and by human synovial tissue lining macrophages.
Blood
1991
, vol. 
78
 
12
(pg. 
3128
-
3132
)
5
McGee
 
MP
Foster
 
S
Wang
 
X
Simultaneous expression of tissue factor and tissue factor pathway inhibitor by human monocytes. A potential mechanism for localized control of blood coagulation.
J Exp Med
1994
, vol. 
179
 
6
(pg. 
1847
-
1854
)
6
Tracy
 
PB
Rohrbach
 
MS
Mann
 
KG
Functional prothrombinase complex assembly on isolated monocytes and lymphocytes.
J Biol Chem
1983
, vol. 
258
 
12
(pg. 
7264
-
7267
)
7
Tracy
 
PB
Eide
 
LL
Mann
 
KG
Human prothrombinase complex assembly and function on isolated peripheral blood cell populations.
J Biol Chem
1985
, vol. 
260
 
4
(pg. 
2119
-
2124
)
8
Butenas
 
S
Bouchard
 
BA
Brummel-Ziedins
 
KE
Parhami-Seren
 
B
Mann
 
KG
Tissue factor activity in whole blood.
Blood
2005
, vol. 
105
 
7
(pg. 
2764
-
2770
)
9
Shantsila
 
E
Lip
 
GYH
The role of monocytes in thrombotic disorders. Insights from tissue factor, monocyte-platelet aggregates and novel mechanisms.
Thromb Haemost
2009
, vol. 
102
 
5
(pg. 
916
-
924
)
10
Ito
 
T
Kawahara
 
K
Nakamura
 
T
, et al. 
High-mobility group box 1 protein promotes development of microvascular thrombosis in rats.
J Thromb Haemost
2007
, vol. 
5
 
1
(pg. 
109
-
116
)
11
Swystun
 
LL
Shin
 
LYY
Beaudin
 
S
Liaw
 
PC
Chemotherapeutic agents doxorubicin and epirubicin induce a procoagulant phenotype on endothelial cells and blood monocytes.
J Thromb Haemost
2009
, vol. 
7
 
4
(pg. 
619
-
626
)
12
Toltl
 
LJ
Beaudin
 
S
Liaw
 
PC
Canadian Critical Care Translational Biology Group
Activated protein C up-regulates IL-10 and inhibits tissue factor in blood monocytes.
J Immunol
2008
, vol. 
181
 
3
(pg. 
2165
-
2173
)
13
Yan
 
SF
Mackman
 
N
Kisiel
 
W
Stern
 
DM
Pinsky
 
DJ
Hypoxia/hypoxemia-induced activation of the procoagulant pathways and the pathogenesis of ischemia-associated thrombosis.
Arterioscler Thromb Vasc Biol
1999
, vol. 
19
 
9
(pg. 
2029
-
2035
)
14
Tutwiler
 
V
Madeeva
 
D
Ahn
 
HS
, et al. 
Platelet transactivation by monocytes promotes thrombosis in heparin-induced thrombocytopenia.
Blood
2016
, vol. 
127
 
4
(pg. 
464
-
472
)
15
Chen
 
VM
Hogg
 
PJ
Encryption and decryption of tissue factor.
J Thromb Haemost
2013
, vol. 
11
 
suppl 1
(pg. 
277
-
284
)
16
Basavaraj
 
MG
Gruber
 
FX
Sovershaev
 
M
, et al. 
The role of TFPI in regulation of TF-induced thrombogenicity on the surface of human monocytes.
Thromb Res
2010
, vol. 
126
 
5
(pg. 
418
-
425
)
17
Aleman
 
MM
Gardiner
 
C
Harrison
 
P
Wolberg
 
AS
Differential contributions of monocyte- and platelet-derived microparticles towards thrombin generation and fibrin formation and stability.
J Thromb Haemost
2011
, vol. 
9
 
11
(pg. 
2251
-
2261
)
18
Angelillo-Scherrer
 
A
Leukocyte-derived microparticles in vascular homeostasis.
Circ Res
2012
, vol. 
110
 
2
(pg. 
356
-
369
)
19
Osterud
 
B
Rao
 
LV
Olsen
 
JO
Induction of tissue factor expression in whole blood: lack of evidence for the presence of tissue factor expression in granulocytes.
Thromb Haemost
2000
, vol. 
83
 
6
(pg. 
861
-
867
)
20
Maugeri
 
N
Brambilla
 
M
Camera
 
M
, et al. 
Human polymorphonuclear leukocytes produce and express functional tissue factor upon stimulation.
J Thromb Haemost
2006
, vol. 
4
 
6
(pg. 
1323
-
1330
)
21
Nakamura
 
S
Imamura
 
T
Okamoto
 
K
Tissue factor in neutrophils: yes.
J Thromb Haemost
2004
, vol. 
2
 
2
(pg. 
214
-
217
)
22
von Brühl
 
M-L
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
, vol. 
209
 
4
(pg. 
819
-
835
)
23
Darbousset
 
R
Thomas
 
GM
Mezouar
 
S
, et al. 
Tissue factor-positive neutrophils bind to injured endothelial wall and initiate thrombus formation.
Blood
2012
, vol. 
120
 
10
(pg. 
2133
-
2143
)
24
Egorina
 
EM
Sovershaev
 
MA
Olsen
 
JO
Østerud
 
B
Granulocytes do not express but acquire monocyte-derived tissue factor in whole blood: evidence for a direct transfer.
Blood
2008
, vol. 
111
 
3
(pg. 
1208
-
1216
)
25
Moosbauer
 
C
Morgenstern
 
E
Cuvelier
 
SL
, et al. 
Eosinophils are a major intravascular location for tissue factor storage and exposure.
Blood
2007
, vol. 
109
 
3
(pg. 
995
-
1002
)
26
Cugno
 
M
Marzano
 
AV
Lorini
 
M
Carbonelli
 
V
Tedeschi
 
A
Enhanced tissue factor expression by blood eosinophils from patients with hypereosinophilia: a possible link with thrombosis.
PLoS One
2014
, vol. 
9
 
11
pg. 
e111862
 
27
Sovershaev
 
MA
Lind
 
KF
Devold
 
H
, et al. 
No evidence for the presence of tissue factor in high-purity preparations of immunologically isolated eosinophils.
J Thromb Haemost
2008
, vol. 
6
 
10
(pg. 
1742
-
1749
)
28
Pham
 
CTN
Neutrophil serine proteases: specific regulators of inflammation.
Nat Rev Immunol
2006
, vol. 
6
 
7
(pg. 
541
-
550
)
29
Meier
 
HL
Heck
 
LW
Schulman
 
ES
MacGlashan
 
DW
Purified human mast cells and basophils release human elastase and cathepsin G by an IgE-mediated mechanism.
Int Arch Allergy Appl Immunol
1985
, vol. 
77
 
1-2
(pg. 
179
-
183
)
30
Allen
 
DH
Tracy
 
PB
Human coagulation factor V is activated to the functional cofactor by elastase and cathepsin G expressed at the monocyte surface.
J Biol Chem
1995
, vol. 
270
 
3
(pg. 
1408
-
1415
)
31
Gale
 
AJ
Rozenshteyn
 
D
Cathepsin G, a leukocyte protease, activates coagulation factor VIII.
Thromb Haemost
2008
, vol. 
99
 
1
(pg. 
44
-
51
)
32
Plescia
 
J
Altieri
 
DC
Activation of Mac-1 (CD11b/CD18)-bound factor X by released cathepsin G defines an alternative pathway of leucocyte initiation of coagulation.
Biochem J
1996
, vol. 
319
 
pt 3
(pg. 
873
-
879
)
33
Jochum
 
M
Lander
 
S
Heimburger
 
N
Fritz
 
H
Effect of human granulocytic elastase on isolated human antithrombin III.
Hoppe Seylers Z Physiol Chem
1981
, vol. 
362
 
2
(pg. 
103
-
112
)
34
Pratt
 
CW
Tobin
 
RB
Church
 
FC
Interaction of heparin cofactor II with neutrophil elastase and cathepsin G.
J Biol Chem
1990
, vol. 
265
 
11
(pg. 
6092
-
6097
)
35
Massberg
 
S
Grahl
 
L
von Bruehl
 
M-L
, et al. 
Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases.
Nat Med
2010
, vol. 
16
 
8
(pg. 
887
-
896
)
36
Belaaouaj
 
AA
Li
 
A
Wun
 
T-C
Welgus
 
HG
Shapiro
 
SD
Matrix metalloproteinases cleave tissue factor pathway inhibitor. Effects on coagulation.
J Biol Chem
2000
, vol. 
275
 
35
(pg. 
27123
-
27128
)
37
Higuchi
 
DA
Wun
 
TC
Likert
 
KM
Broze
 
GJ
The effect of leukocyte elastase on tissue factor pathway inhibitor.
Blood
1992
, vol. 
79
 
7
(pg. 
1712
-
1719
)
38
Maugeri
 
N
Campana
 
L
Gavina
 
M
, et al. 
Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps.
J Thromb Haemost
2014
, vol. 
12
 
12
(pg. 
2074
-
2088
)
39
Kaplan
 
MJ
Radic
 
M
Neutrophil extracellular traps: double-edged swords of innate immunity.
J Immunol
2012
, vol. 
189
 
6
(pg. 
2689
-
2695
)
40
Tadie
 
J-M
Bae
 
H-B
Jiang
 
S
, et al. 
HMGB1 promotes neutrophil extracellular trap formation through interactions with toll-like receptor 4.
Am J Physiol Lung Cell Mol Physiol
2013
, vol. 
304
 
5
(pg. 
L342
-
L349
)
41
Clark
 
SR
Ma
 
AC
Tavener
 
SA
, et al. 
Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood.
Nat Med
2007
, vol. 
13
 
4
(pg. 
463
-
469
)
42
Chow
 
OA
von Köckritz-Blickwede
 
M
Bright
 
AT
, et al. 
Statins enhance formation of phagocyte extracellular traps.
Cell Host Microbe
2010
, vol. 
8
 
5
(pg. 
445
-
454
)
43
von Köckritz-Blickwede
 
M
Goldmann
 
O
Thulin
 
P
, et al. 
Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation.
Blood
2008
, vol. 
111
 
6
(pg. 
3070
-
3080
)
44
Morshed
 
M
Hlushchuk
 
R
Simon
 
D
, et al. 
NADPH oxidase-independent formation of extracellular DNA traps by basophils.
J Immunol
2014
, vol. 
192
 
11
(pg. 
5314
-
5323
)
45
Yousefi
 
S
Gold
 
JA
Andina
 
N
, et al. 
Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense.
Nat Med
2008
, vol. 
14
 
9
(pg. 
949
-
953
)
46
Jahr
 
S
Hentze
 
H
Englisch
 
S
, et al. 
DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells.
Cancer Res
2001
, vol. 
61
 
4
(pg. 
1659
-
1665
)
47
Fuchs
 
TA
Brill
 
A
Wagner
 
DD
Neutrophil extracellular trap (NET) impact on deep vein thrombosis.
Arterioscler Thromb Vasc Biol
2012
, vol. 
32
 
8
(pg. 
1777
-
1783
)
48
Mitroulis
 
I
Kambas
 
K
Chrysanthopoulou
 
A
, et al. 
Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout.
PLoS One
2011
, vol. 
6
 
12
pg. 
e29318
 
49
Swystun
 
LL
Mukherjee
 
S
Liaw
 
PC
Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus.
J Thromb Haemost
2011
, vol. 
9
 
11
(pg. 
2313
-
2321
)
50
Gould
 
TJ
Vu
 
TT
Swystun
 
LL
, et al. 
Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms.
Arterioscler Thromb Vasc Biol
2014
, vol. 
34
 
9
(pg. 
1977
-
1984
)
51
Barranco-Medina
 
S
Pozzi
 
N
Vogt
 
AD
Di Cera
 
E
Histone H4 promotes prothrombin autoactivation.
J Biol Chem
2013
, vol. 
288
 
50
(pg. 
35749
-
35757
)
52
Longstaff
 
C
Hogwood
 
J
Gray
 
E
, et al. 
Neutralisation of the anti-coagulant effects of heparin by histones in blood plasma and purified systems.
Thromb Haemost
2016
, vol. 
115
 
3
(pg. 
591
-
599
)
53
Varjú
 
I
Longstaff
 
C
Szabó
 
L
, et al. 
DNA, histones and neutrophil extracellular traps exert anti-fibrinolytic effects in a plasma environment.
Thromb Haemost
2015
, vol. 
113
 
6
(pg. 
1289
-
1298
)
54
Ammollo
 
CT
Semeraro
 
F
Xu
 
J
Esmon
 
NL
Esmon
 
CT
Extracellular histones increase plasma thrombin generation by impairing thrombomodulin-dependent protein C activation.
J Thromb Haemost
2011
, vol. 
9
 
9
(pg. 
1795
-
1803
)
55
Nan
 
B
Lin
 
P
Lumsden
 
AB
Yao
 
Q
Chen
 
C
Effects of TNF-alpha and curcumin on the expression of thrombomodulin and endothelial protein C receptor in human endothelial cells.
Thromb Res
2005
, vol. 
115
 
5
(pg. 
417
-
426
)
56
Menschikowski
 
M
Hagelgans
 
A
Eisenhofer
 
G
Siegert
 
G
Regulation of endothelial protein C receptor shedding by cytokines is mediated through differential activation of MAP kinase signaling pathways.
Exp Cell Res
2009
, vol. 
315
 
15
(pg. 
2673
-
2682
)
57
Xu
 
J
Zhang
 
X
Pelayo
 
R
, et al. 
Extracellular histones are major mediators of death in sepsis.
Nat Med
2009
, vol. 
15
 
11
(pg. 
1318
-
1321
)
58
Semeraro
 
F
Ammollo
 
CT
Esmon
 
NL
Esmon
 
CT
Histones induce phosphatidylserine exposure and a procoagulant phenotype in human red blood cells.
J Thromb Haemost
2014
, vol. 
12
 
10
(pg. 
1697
-
1702
)
59
Yang
 
X
Li
 
L
Liu
 
J
Lv
 
B
Chen
 
F
Extracellular histones induce tissue factor expression in vascular endothelial cells via TLR and activation of NF-κB and AP-1.
Thromb Res
2016
, vol. 
137
 (pg. 
211
-
218
)
60
Herbert
 
JM
Savi
 
P
Laplace
 
MC
Lale
 
A
IL-4 inhibits LPS-, IL-1 beta- and TNF alpha-induced expression of tissue factor in endothelial cells and monocytes.
FEBS Lett
1992
, vol. 
310
 
1
(pg. 
31
-
33
)
61
Haubitz
 
M
Gerlach
 
M
Kruse
 
HJ
Brunkhorst
 
R
Endothelial tissue factor stimulation by proteinase 3 and elastase.
Clin Exp Immunol
2001
, vol. 
126
 
3
(pg. 
584
-
588
)
62
Michels
 
A
Swystun
 
LL
Albánez
 
S
, et al. 
Histones induce endothelial von Willebrand factor release and subsequent platelet capture in in vitro and in vivo models [abstract].
Blood
2014
, vol. 
123
 
21
 
Abstract 2768
63
Bernardo
 
A
Ball
 
C
Nolasco
 
L
Moake
 
JF
Dong
 
JF
Effects of inflammatory cytokines on the release and cleavage of the endothelial cell-derived ultralarge von Willebrand factor multimers under flow.
Blood
2004
, vol. 
104
 
1
(pg. 
100
-
106
)
64
Hamilton
 
KK
Sims
 
PJ
Changes in cytosolic Ca2+ associated with von Willebrand factor release in human endothelial cells exposed to histamine. Study of microcarrier cell monolayers using the fluorescent probe indo-1.
J Clin Invest
1987
, vol. 
79
 
2
(pg. 
600
-
608
)
65
Chen
 
J
Fu
 
X
Wang
 
Y
, et al. 
Oxidative modification of von Willebrand factor by neutrophil oxidants inhibits its cleavage by ADAMTS13.
Blood
2010
, vol. 
115
 
3
(pg. 
706
-
712
)
66
Yan
 
SLS
Russell
 
J
Granger
 
DN
Platelet activation and platelet-leukocyte aggregation elicited in experimental colitis are mediated by interleukin-6.
Inflamm Bowel Dis
2014
, vol. 
20
 
2
(pg. 
353
-
362
)
67
Christersson
 
C
Johnell
 
M
Siegbahn
 
A
The influence of direct thrombin inhibitors on the formation of platelet-leukocyte aggregates and tissue factor expression.
Thromb Res
2010
, vol. 
126
 
4
(pg. 
e327
-
e333
)
68
Hu
 
H
Varon
 
D
Hjemdahl
 
P
Savion
 
N
Schulman
 
S
Li
 
N
Platelet-leukocyte aggregation under shear stress: differential involvement of selectins and integrins.
Thromb Haemost
2003
, vol. 
90
 
4
(pg. 
679
-
687
)
69
Carestia
 
A
Rivadeneyra
 
L
Romaniuk
 
MA
Fondevila
 
C
Negrotto
 
S
Schattner
 
M
Functional responses and molecular mechanisms involved in histone-mediated platelet activation.
Thromb Haemost
2013
, vol. 
110
 
5
(pg. 
1035
-
1045
)
70
Rinder
 
HM
Bonan
 
JL
Rinder
 
CS
Ault
 
KA
Smith
 
BR
Dynamics of leukocyte-platelet adhesion in whole blood.
Blood
1991
, vol. 
78
 
7
(pg. 
1730
-
1737
)
71
Diacovo
 
TG
Roth
 
SJ
Buccola
 
JM
Bainton
 
DF
Springer
 
TA
Neutrophil rolling, arrest, and transmigration across activated, surface-adherent platelets via sequential action of P-selectin and the beta 2-integrin CD11b/CD18.
Blood
1996
, vol. 
88
 
1
(pg. 
146
-
157
)
72
Simon
 
DI
Chen
 
Z
Xu
 
H
, et al. 
Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18).
J Exp Med
2000
, vol. 
192
 
2
(pg. 
193
-
204
)
73
Weber
 
C
Springer
 
TA
Neutrophil accumulation on activated, surface-adherent platelets in flow is mediated by interaction of Mac-1 with fibrinogen bound to alphaIIbbeta3 and stimulated by platelet-activating factor.
J Clin Invest
1997
, vol. 
100
 
8
(pg. 
2085
-
2093
)
74
Lievens
 
D
Zernecke
 
A
Seijkens
 
T
, et al. 
Platelet CD40L mediates thrombotic and inflammatory processes in atherosclerosis.
Blood
2010
, vol. 
116
 
20
(pg. 
4317
-
4327
)
75
Schulz
 
C
von Brühl
 
ML
Barocke
 
V
, et al. 
EMMPRIN (CD147/basigin) mediates platelet-monocyte interactions in vivo and augments monocyte recruitment to the vascular wall.
J Thromb Haemost
2011
, vol. 
9
 
5
(pg. 
1007
-
1019
)
76
Diacovo
 
TG
deFougerolles
 
AR
Bainton
 
DF
Springer
 
TA
A functional integrin ligand on the surface of platelets: intercellular adhesion molecule-2.
J Clin Invest
1994
, vol. 
94
 
3
(pg. 
1243
-
1251
)
77
Santoso
 
S
Sachs
 
UJH
Kroll
 
H
, et al. 
The junctional adhesion molecule 3 (JAM-3) on human platelets is a counterreceptor for the leukocyte integrin Mac-1.
J Exp Med
2002
, vol. 
196
 
5
(pg. 
679
-
691
)
78
Weyrich
 
AS
Elstad
 
MR
McEver
 
RP
, et al. 
Activated platelets signal chemokine synthesis by human monocytes.
J Clin Invest
1996
, vol. 
97
 
6
(pg. 
1525
-
1534
)
79
Celi
 
A
Pellegrini
 
G
Lorenzet
 
R
, et al. 
P-selectin induces the expression of tissue factor on monocytes.
Proc Natl Acad Sci USA
1994
, vol. 
91
 
19
(pg. 
8767
-
8771
)
80
Renesto
 
P
Chignard
 
M
Enhancement of cathepsin G-induced platelet activation by leukocyte elastase: consequence for the neutrophil-mediated platelet activation.
Blood
1993
, vol. 
82
 
1
(pg. 
139
-
144
)
81
LaRosa
 
CA
Rohrer
 
MJ
Benoit
 
SE
Rodino
 
LJ
Barnard
 
MR
Michelson
 
AD
Human neutrophil cathepsin G is a potent platelet activator.
J Vasc Surg
1994
, vol. 
19
 
2
(pg. 
306
-
318, discussion 318-319
)
82
Seth
 
P
Kumari
 
R
Dikshit
 
M
Srimal
 
RC
Effect of platelet activating factor antagonists in different models of thrombosis.
Thromb Res
1994
, vol. 
76
 
6
(pg. 
503
-
512
)
83
Fuchs
 
TA
Bhandari
 
AA
Wagner
 
DD
Histones induce rapid and profound thrombocytopenia in mice.
Blood
2011
, vol. 
118
 
13
(pg. 
3708
-
3714
)
84
Semeraro
 
F
Ammollo
 
CT
Morrissey
 
JH
, et al. 
Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4.
Blood
2011
, vol. 
118
 
7
(pg. 
1952
-
1961
)
85
Diaz
 
JA
Fuchs
 
TA
Jackson
 
TO
, et al. 
for the Michigan Research Venous Group*
Plasma DNA is elevated in patients with deep vein thrombosis.
J Vasc Surg Venous Lymphat Disord
2013
, vol. 
1
 
4
(pg. 
341
-
348
)
86
van Montfoort
 
ML
Stephan
 
F
Lauw
 
MN
, et al. 
Circulating nucleosomes and neutrophil activation as risk factors for deep vein thrombosis.
Arterioscler Thromb Vasc Biol
2013
, vol. 
33
 
1
(pg. 
147
-
151
)
87
Hölschermann
 
H
Haberbosch
 
W
Terhalle
 
H-M
, et al. 
Increased monocyte tissue factor activity in women following cerebral venous thrombosis.
J Neurol
2003
, vol. 
250
 
5
(pg. 
631
-
632
)
88
Kamikura
 
Y
Wada
 
H
Nobori
 
T
, et al. 
Elevated levels of leukocyte tissue factor mRNA in patients with venous thromboembolism.
Thromb Res
2005
, vol. 
116
 
4
(pg. 
307
-
312
)
89
Himber
 
J
Kling
 
D
Fallon
 
JT
Nemerson
 
Y
Riederer
 
MA
In situ localization of tissue factor in human thrombi.
Blood
2002
, vol. 
99
 
11
(pg. 
4249
-
4250
)
90
Savchenko
 
AS
Martinod
 
K
Seidman
 
MA
, et al. 
Neutrophil extracellular traps form predominantly during the organizing stage of human venous thromboembolism development.
J Thromb Haemost
2014
, vol. 
12
 
6
(pg. 
860
-
870
)
91
Semple
 
JW
Italiano
 
JE
Freedman
 
J
Platelets and the immune continuum.
Nat Rev Immunol
2011
, vol. 
11
 
4
(pg. 
264
-
274
)
92
Kaplan
 
ZS
Zarpellon
 
A
Alwis
 
I
, et al. 
Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4.
Nat Commun
2015
, vol. 
6
 pg. 
7835
 
93
Sullivan
 
VV
Hawley
 
AE
Farris
 
DM
, et al. 
Decrease in fibrin content of venous thrombi in selectin-deficient mice.
J Surg Res
2003
, vol. 
109
 
1
(pg. 
1
-
7
)
94
Pendu
 
R
Terraube
 
V
Christophe
 
OD
, et al. 
P-selectin glycoprotein ligand 1 and beta2-integrins cooperate in the adhesion of leukocytes to von Willebrand factor.
Blood
2006
, vol. 
108
 
12
(pg. 
3746
-
3752
)
95
Zarbock
 
A
Ley
 
K
McEver
 
RP
Hidalgo
 
A
Leukocyte ligands for endothelial selectins: specialized glycoconjugates that mediate rolling and signaling under flow.
Blood
2011
, vol. 
118
 
26
(pg. 
6743
-
6751
)
96
Rao
 
RM
Yang
 
L
Garcia-Cardena
 
G
Luscinskas
 
FW
Endothelial-dependent mechanisms of leukocyte recruitment to the vascular wall.
Circ Res
2007
, vol. 
101
 
3
(pg. 
234
-
247
)
97
Laurance
 
S
Bertin
 
F-R
Ebrahimian
 
T
, et al. 
Gas6 promotes pro-inflammatory (Ly6Chi) monocyte recruitment in venous thrombosis.
Blood
2014
, vol. 
124
 
21
pg. 
1533
 
98
Niemetz
 
J
Fani
 
K
Thrombogenic activity of leukocytes.
Blood
1973
, vol. 
42
 
1
(pg. 
47
-
59
)
99
Himber
 
J
Wohlgensinger
 
C
Roux
 
S
, et al. 
Inhibition of tissue factor limits the growth of venous thrombus in the rabbit.
J Thromb Haemost
2003
, vol. 
1
 
5
(pg. 
889
-
895
)
100
Zhou
 
J
May
 
L
Liao
 
P
Gross
 
PL
Weitz
 
JI
Inferior vena cava ligation rapidly induces tissue factor expression and venous thrombosis in rats.
Arterioscler Thromb Vasc Biol
2009
, vol. 
29
 
6
(pg. 
863
-
869
)
101
Day
 
SM
Reeve
 
JL
Pedersen
 
B
, et al. 
Macrovascular thrombosis is driven by tissue factor derived primarily from the blood vessel wall.
Blood
2005
, vol. 
105
 
1
(pg. 
192
-
198
)
102
Fuchs
 
TA
Brill
 
A
Duerschmied
 
D
, et al. 
Extracellular DNA traps promote thrombosis.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
36
(pg. 
15880
-
15885
)
103
Brill
 
A
Fuchs
 
TA
Savchenko
 
AS
, et al. 
Neutrophil extracellular traps promote deep vein thrombosis in mice.
J Thromb Haemost
2012
, vol. 
10
 
1
(pg. 
136
-
144
)
104
Heestermans
 
M
Salloum-Asfar
 
S
Salvatori
 
D
, et al. 
Role of platelets, neutrophils, and factor XII in spontaneous venous thrombosis in mice.
Blood
2016
, vol. 
127
 
21
(pg. 
2630
-
2637
)
105
Martinod
 
K
Demers
 
M
Fuchs
 
TA
, et al. 
Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
21
(pg. 
8674
-
8679
)
106
El-Sayed
 
OM
Dewyer
 
NA
Luke
 
CE
, et al. 
Intact toll-like receptor 9 signaling in neutrophils modulates normal thrombogenesis in mice [published online ahead of print October 16, 2015].
J Vasc Surg
 
doi:10.1016/j.jvs.2015.08.070
107
Papayannopoulos
 
V
Metzler
 
KD
Hakkim
 
A
Zychlinsky
 
A
Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps.
J Cell Biol
2010
, vol. 
191
 
3
(pg. 
677
-
691
)
108
Kolaczkowska
 
E
Jenne
 
CN
Surewaard
 
BGJ
, et al. 
Molecular mechanisms of NET formation and degradation revealed by intravital imaging in the liver vasculature.
Nat Commun
2015
, vol. 
6
 pg. 
6673
 
109
Martinod
 
K
Witsch
 
T
Farley
 
K
Gallant
 
M
Remold-O’Donnell
 
E
Wagner
 
DD
Neutrophil elastase-deficient mice form neutrophil extracellular traps in an experimental model of deep vein thrombosis.
J Thromb Haemost
2016
, vol. 
14
 
3
(pg. 
551
-
558
)
110
Wilcox
 
JN
Smith
 
KM
Schwartz
 
SM
Gordon
 
D
Localization of tissue factor in the normal vessel wall and in the atherosclerotic plaque.
Proc Natl Acad Sci USA
1989
, vol. 
86
 
8
(pg. 
2839
-
2843
)
111
Mallat
 
Z
Hugel
 
B
Ohan
 
J
Lesèche
 
G
Freyssinet
 
JM
Tedgui
 
A
Shed membrane microparticles with procoagulant potential in human atherosclerotic plaques: a role for apoptosis in plaque thrombogenicity.
Circulation
1999
, vol. 
99
 
3
(pg. 
348
-
353
)
112
Lindemann
 
S
Tolley
 
ND
Dixon
 
DA
, et al. 
Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis.
J Cell Biol
2001
, vol. 
154
 
3
(pg. 
485
-
490
)
113
Owens
 
AP
Passam
 
FH
Antoniak
 
S
, et al. 
Monocyte tissue factor-dependent activation of coagulation in hypercholesterolemic mice and monkeys is inhibited by simvastatin.
J Clin Invest
2012
, vol. 
122
 
2
(pg. 
558
-
568
)
114
Borissoff
 
JI
Joosen
 
IA
Versteylen
 
MO
, et al. 
Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state.
Arterioscler Thromb Vasc Biol
2013
, vol. 
33
 
8
(pg. 
2032
-
2040
)
115
Ramaiola
 
I
Padró
 
T
Peña
 
E
, et al. 
Changes in thrombus composition and profilin-1 release in acute myocardial infarction.
Eur Heart J
2015
, vol. 
36
 
16
(pg. 
965
-
975
)
116
Riegger
 
J
Byrne
 
RA
Joner
 
M
, et al. 
Prevention of Late Stent Thrombosis by an Interdisciplinary Global European Effort (PRESTIGE) Investigators
Histopathological evaluation of thrombus in patients presenting with stent thrombosis. A multicenter European study: a report of the prevention of late stent thrombosis by an interdisciplinary global European effort consortium.
Eur Heart J
2016
, vol. 
37
 
19
(pg. 
1538
-
1549
)
117
Chi
 
L
Gibson
 
G
Peng
 
Y-W
, et al. 
Characterization of a tissue factor/factor VIIa-dependent model of thrombosis in hypercholesterolemic rabbits.
J Thromb Haemost
2004
, vol. 
2
 
1
(pg. 
85
-
92
)
118
Palabrica
 
T
Lobb
 
R
Furie
 
BC
, et al. 
Leukocyte accumulation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets.
Nature
1992
, vol. 
359
 
6398
(pg. 
848
-
851
)
119
Reinhardt
 
C
von Brühl
 
ML
Manukyan
 
D
, et al. 
Protein disulfide isomerase acts as an injury response signal that enhances fibrin generation via tissue factor activation.
J Clin Invest
2008
, vol. 
118
 
3
(pg. 
1110
-
1122
)
120
Megens
 
RT
Vijayan
 
S
Lievens
 
D
, et al. 
Presence of luminal neutrophil extracellular traps in atherosclerosis.
Thromb Haemost
2012
, vol. 
107
 
3
(pg. 
597
-
598
)
121
Stakos
 
DA
Kambas
 
K
Konstantinidis
 
T
, et al. 
Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction.
Eur Heart J
2015
, vol. 
36
 
22
(pg. 
1405
-
1414
)
122
Kannemeier
 
C
Shibamiya
 
A
Nakazawa
 
F
, et al. 
Extracellular RNA constitutes a natural procoagulant cofactor in blood coagulation.
Proc Natl Acad Sci USA
2007
, vol. 
104
 
15
(pg. 
6388
-
6393
)
123
Knight
 
JS
Zhao
 
W
Luo
 
W
, et al. 
Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus.
J Clin Invest
2013
, vol. 
123
 
7
(pg. 
2981
-
2993
)
124
Knight
 
JS
Luo
 
W
O’Dell
 
AA
, et al. 
Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis.
Circ Res
2014
, vol. 
114
 
6
(pg. 
947
-
956
)
125
Fuchs
 
TA
Kremer Hovinga
 
JA
Schatzberg
 
D
Wagner
 
DD
Lämmle
 
B
Circulating DNA and myeloperoxidase indicate disease activity in patients with thrombotic microangiopathies.
Blood
2012
, vol. 
120
 
6
(pg. 
1157
-
1164
)
126
Jiménez-Alcázar
 
M
Napirei
 
M
Panda
 
R
, et al. 
Impaired DNase1-mediated degradation of neutrophil extracellular traps is associated with acute thrombotic microangiopathies.
J Thromb Haemost
2015
, vol. 
13
 
5
(pg. 
732
-
742
)
127
Falati
 
S
Liu
 
Q
Gross
 
P
, et al. 
Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin.
J Exp Med
2003
, vol. 
197
 
11
(pg. 
1585
-
1598
)
128
Chou
 
J
Mackman
 
N
Merrill-Skoloff
 
G
Pedersen
 
B
Furie
 
BC
Furie
 
B
Hematopoietic cell-derived microparticle tissue factor contributes to fibrin formation during thrombus propagation.
Blood
2004
, vol. 
104
 
10
(pg. 
3190
-
3197
)
129
Gross
 
PL
Furie
 
BC
Merrill-Skoloff
 
G
Chou
 
J
Furie
 
B
Leukocyte-versus microparticle-mediated tissue factor transfer during arteriolar thrombus development.
J Leukoc Biol
2005
, vol. 
78
 
6
(pg. 
1318
-
1326
)
130
Bancsi
 
MJ
Thompson
 
J
Bertina
 
RM
Stimulation of monocyte tissue factor expression in an in vitro model of bacterial endocarditis.
Infect Immun
1994
, vol. 
62
 
12
(pg. 
5669
-
5672
)
131
Guha
 
M
O’Connell
 
MA
Pawlinski
 
R
, et al. 
Lipopolysaccharide activation of the MEK-ERK1/2 pathway in human monocytic cells mediates tissue factor and tumor necrosis factor alpha expression by inducing Elk-1 phosphorylation and Egr-1 expression.
Blood
2001
, vol. 
98
 
5
(pg. 
1429
-
1439
)
132
Wen
 
B
Combes
 
V
Bonhoure
 
A
Weksler
 
BB
Couraud
 
PO
Grau
 
GE
Endotoxin-induced monocytic microparticles have contrasting effects on endothelial inflammatory responses.
PLoS One
2014
, vol. 
9
 
3
pg. 
e91597
 
133
Vickers
 
J
Russwurm
 
S
Dohrn
 
B
, et al. 
Monocyte tissue factor (CD142) and Mac-1 (CD11b) are increased in septic patients.
Thromb Haemost
1998
, vol. 
79
 
6
(pg. 
1219
-
1220
)
134
Hellum
 
M
Øvstebø
 
R
Brusletto
 
BS
Berg
 
JP
Brandtzaeg
 
P
Henriksson
 
CE
Microparticle-associated tissue factor activity correlates with plasma levels of bacterial lipopolysaccharides in meningococcal septic shock.
Thromb Res
2014
, vol. 
133
 
3
(pg. 
507
-
514
)
135
Sundén-Cullberg
 
J
Norrby-Teglund
 
A
Rouhiainen
 
A
, et al. 
Persistent elevation of high mobility group box-1 protein (HMGB1) in patients with severe sepsis and septic shock.
Crit Care Med
2005
, vol. 
33
 
3
(pg. 
564
-
573
)
136
Dwivedi
 
DJ
Toltl
 
LJ
Swystun
 
LL
, et al. 
Canadian Critical Care Translational Biology Group
Prognostic utility and characterization of cell-free DNA in patients with severe sepsis.
Crit Care
2012
, vol. 
16
 
4
pg. 
R151
 
137
Rondina
 
MT
Carlisle
 
M
Fraughton
 
T
, et al. 
Platelet-monocyte aggregate formation and mortality risk in older patients with severe sepsis and septic shock.
J Gerontol A Biol Sci Med Sci
2015
, vol. 
70
 
2
(pg. 
225
-
231
)
138
Pawlinski
 
R
Wang
 
JG
Owens
 
AP
, et al. 
Hematopoietic and nonhematopoietic cell tissue factor activates the coagulation cascade in endotoxemic mice.
Blood
2010
, vol. 
116
 
5
(pg. 
806
-
814
)
139
Adams
 
SA
Kelly
 
SL
Kirsch
 
RE
Robson
 
SC
Shephard
 
EG
Role of neutrophil membrane proteases in fibrin degradation.
Blood Coagul Fibrinolysis
1995
, vol. 
6
 
8
(pg. 
693
-
702
)
140
Grau
 
E
Moroz
 
LA
Fibrinolytic activity of normal human blood monocytes.
Thromb Res
1989
, vol. 
53
 
2
(pg. 
145
-
162
)
141
Lacroix
 
R
Plawinski
 
L
Robert
 
S
, et al. 
Leukocyte- and endothelial-derived microparticles: a circulating source for fibrinolysis.
Haematologica
2012
, vol. 
97
 
12
(pg. 
1864
-
1872
)
142
Kunigal
 
S
Kusch
 
A
Tkachuk
 
N
, et al. 
Monocyte-expressed urokinase inhibits vascular smooth muscle cell growth by activating Stat1.
Blood
2003
, vol. 
102
 
13
(pg. 
4377
-
4383
)
143
Singh
 
I
Burnand
 
KG
Collins
 
M
, et al. 
Failure of thrombus to resolve in urokinase-type plasminogen activator gene-knockout mice: rescue by normal bone marrow-derived cells.
Circulation
2003
, vol. 
107
 
6
(pg. 
869
-
875
)
144
Das
 
R
Pluskota
 
E
Plow
 
EF
Plasminogen and its receptors as regulators of cardiovascular inflammatory responses.
Trends Cardiovasc Med
2010
, vol. 
20
 
4
(pg. 
120
-
124
)
145
Wu
 
K
Urano
 
T
Ihara
 
H
, et al. 
The cleavage and inactivation of plasminogen activator inhibitor type 1 by neutrophil elastase: the evaluation of its physiologic relevance in fibrinolysis.
Blood
1995
, vol. 
86
 
3
(pg. 
1056
-
1061
)
146
Machovich
 
R
Himer
 
A
Owen
 
WG
Neutrophil proteases in plasminogen activation.
Blood Coagul Fibrinolysis
1990
, vol. 
1
 
3
(pg. 
273
-
277
)
147
Semeraro
 
F
Ammollo
 
CT
Semeraro
 
N
Colucci
 
M
Tissue factor-expressing monocytes inhibit fibrinolysis through a TAFI-mediated mechanism, and make clots resistant to heparins.
Haematologica
2009
, vol. 
94
 
6
(pg. 
819
-
826
)
148
Longstaff
 
C
Varjú
 
I
Sótonyi
 
P
, et al. 
Mechanical stability and fibrinolytic resistance of clots containing fibrin, DNA, and histones.
J Biol Chem
2013
, vol. 
288
 
10
(pg. 
6946
-
6956
)
149
Gould
 
TJ
Vu
 
TT
Stafford
 
AR
, et al. 
Cell-free DNA modulates clot structure and impairs fibrinolysis in sepsis.
Arterioscler Thromb Vasc Biol
2015
, vol. 
35
 
12
(pg. 
2544
-
2553
)
150
Komissarov
 
AA
Florova
 
G
Idell
 
S
Effects of extracellular DNA on plasminogen activation and fibrinolysis.
J Biol Chem
2011
, vol. 
286
 
49
(pg. 
41949
-
41962
)
151
van Schooten
 
CJ
Shahbazi
 
S
Groot
 
E
, et al. 
Macrophages contribute to the cellular uptake of von Willebrand factor and factor VIII in vivo.
Blood
2008
, vol. 
112
 
5
(pg. 
1704
-
1712
)
152
Pegon
 
JN
Kurdi
 
M
Casari
 
C
, et al. 
Factor VIII and von Willebrand factor are ligands for the carbohydrate-receptor Siglec-5.
Haematologica
2012
, vol. 
97
 
12
(pg. 
1855
-
1863
)
153
Hoffmeister
 
KM
The role of lectins and glycans in platelet clearance.
J Thromb Haemost
2011
, vol. 
9
 
supp 1
(pg. 
35
-
43
)
154
Simon
 
DI
Ezratty
 
AM
Francis
 
SA
Rennke
 
H
Loscalzo
 
J
Fibrin(ogen) is internalized and degraded by activated human monocytoid cells via Mac-1 (CD11b/CD18): a nonplasmin fibrinolytic pathway.
Blood
1993
, vol. 
82
 
8
(pg. 
2414
-
2422
)
155
Farrera
 
C
Fadeel
 
B
Macrophage clearance of neutrophil extracellular traps is a silent process.
J Immunol
2013
, vol. 
191
 
5
(pg. 
2647
-
2656
)
156
Maugeri
 
N
Rovere-Querini
 
P
Evangelista
 
V
, et al. 
Neutrophils phagocytose activated platelets in vivo: a phosphatidylserine, P-selectin, and beta2 integrin-dependent cell clearance program.
Blood
2009
, vol. 
113
 
21
(pg. 
5254
-
5265
)
157
Varma
 
MR
Varga
 
AJ
Knipp
 
BS
, et al. 
Neutropenia impairs venous thrombosis resolution in the rat.
J Vasc Surg
2003
, vol. 
38
 
5
(pg. 
1090
-
1098
)
158
Henke
 
PK
Pearce
 
CG
Moaveni
 
DM
, et al. 
Targeted deletion of CCR2 impairs deep vein thombosis resolution in a mouse model.
J Immunol
2006
, vol. 
177
 
5
(pg. 
3388
-
3397
)
159
Wakefield
 
TW
Strieter
 
RM
Wilke
 
CA
, et al. 
Venous thrombosis-associated inflammation and attenuation with neutralizing antibodies to cytokines and adhesion molecules.
Arterioscler Thromb Vasc Biol
1995
, vol. 
15
 
2
(pg. 
258
-
268
)
160
Smiley
 
ST
King
 
JA
Hancock
 
WW
Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4.
J Immunol
2001
, vol. 
167
 
5
(pg. 
2887
-
2894
)
161
Perez
 
RL
Roman
 
J
Fibrin enhances the expression of IL-1 beta by human peripheral blood mononuclear cells. Implications in pulmonary inflammation.
J Immunol
1995
, vol. 
154
 
4
(pg. 
1879
-
1887
)
162
Rubel
 
C
Fernández
 
GC
Dran
 
G
Bompadre
 
MB
Isturiz
 
MA
Palermo
 
MS
Fibrinogen promotes neutrophil activation and delays apoptosis.
J Immunol
2001
, vol. 
166
 
3
(pg. 
2002
-
2010
)
163
Henke
 
PK
Varga
 
A
De
 
S
, et al. 
Deep vein thrombosis resolution is modulated by monocyte CXCR2-mediated activity in a mouse model.
Arterioscler Thromb Vasc Biol
2004
, vol. 
24
 
6
(pg. 
1130
-
1137
)
164
Soo
 
KS
Northeast
 
AD
Happerfield
 
LC
Burnand
 
KG
Bobrow
 
LG
Tissue plasminogen activator production by monocytes in venous thrombolysis.
J Pathol
1996
, vol. 
178
 
2
(pg. 
190
-
194
)
165
Wakefield
 
TW
Myers
 
DD
Henke
 
PK
Mechanisms of venous thrombosis and resolution.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 
3
(pg. 
387
-
391
)
166
Gordon
 
S
Taylor
 
PR
Monocyte and macrophage heterogeneity.
Nat Rev Immunol
2005
, vol. 
5
 
12
(pg. 
953
-
964
)
167
Howells
 
GL
Macey
 
MG
Chinni
 
C
, et al. 
Proteinase-activated receptor-2: expression by human neutrophils.
J Cell Sci
1997
, vol. 
110
 
pt 7
(pg. 
881
-
887
)
168
Bizios
 
R
Lai
 
L
Fenton
 
JW
Malik
 
AB
Thrombin-induced chemotaxis and aggregation of neutrophils.
J Cell Physiol
1986
, vol. 
128
 
3
(pg. 
485
-
490
)
169
Bar-Shavit
 
R
Kahn
 
A
Fenton
 
JW
Wilner
 
GD
Chemotactic response of monocytes to thrombin.
J Cell Biol
1983
, vol. 
96
 
1
(pg. 
282
-
285
)
170
Jenkins
 
AL
Howells
 
GL
Scott
 
E
Le Bonniec
 
BF
Curtis
 
MA
Stone
 
SR
The response to thrombin of human neutrophils: evidence for two novel receptors.
J Cell Sci
1995
, vol. 
108
 
pt 9
(pg. 
3059
-
3066
)
171
Stephenson
 
DA
Toltl
 
LJ
Beaudin
 
S
Liaw
 
PC
Modulation of monocyte function by activated protein C, a natural anticoagulant.
J Immunol
2006
, vol. 
177
 
4
(pg. 
2115
-
2122
)
172
Elphick
 
GF
Sarangi
 
PP
Hyun
 
Y-M
, et al. 
Recombinant human activated protein C inhibits integrin-mediated neutrophil migration.
Blood
2009
, vol. 
113
 
17
(pg. 
4078
-
4085
)
173
Naldini
 
A
Sower
 
L
Bocci
 
V
Meyers
 
B
Carney
 
DH
Thrombin receptor expression and responsiveness of human monocytic cells to thrombin is linked to interferon-induced cellular differentiation.
J Cell Physiol
1998
, vol. 
177
 
1
(pg. 
76
-
84
)
174
Totani
 
L
Amore
 
C
Di Santo
 
A
, et al. 
Roflumilast inhibits leukocyte-platelet interactions and prevents the prothrombotic functions of polymorphonuclear leukocytes and monocytes.
J Thromb Haemost
2016
, vol. 
14
 
1
(pg. 
191
-
204
)
175
White
 
WB
Cooke
 
GE
Kowey
 
PR
, et al. 
Cardiovascular safety in patients receiving roflumilast for the treatment of COPD.
Chest
2013
, vol. 
144
 
3
(pg. 
758
-
765
)
176
Ridker
 
PM
Danielson
 
E
Fonseca
 
FAH
, et al. 
JUPITER Study Group
Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein.
N Engl J Med
2008
, vol. 
359
 
21
(pg. 
2195
-
2207
)
177
Glynn
 
RJ
Danielson
 
E
Fonseca
 
FAH
, et al. 
A randomized trial of rosuvastatin in the prevention of venous thromboembolism.
N Engl J Med
2009
, vol. 
360
 
18
(pg. 
1851
-
1861
)
178
Downing
 
LJ
Wakefield
 
TW
Strieter
 
RM
, et al. 
Anti-P-selectin antibody decreases inflammation and thrombus formation in venous thrombosis.
J Vasc Surg
1997
, vol. 
25
 
5
(pg. 
816
-
827, discussion 828
)
179
Ramacciotti
 
E
Myers
 
DD
Wrobleski
 
SK
, et al. 
P-selectin/ PSGL-1 inhibitors versus enoxaparin in the resolution of venous thrombosis: a meta-analysis.
Thromb Res
2010
, vol. 
125
 
4
(pg. 
e138
-
e142
)
180
Nakahara
 
M
Ito
 
T
Kawahara
 
K
, et al. 
Recombinant thrombomodulin protects mice against histone-induced lethal thromboembolism.
PLoS One
2013
, vol. 
8
 
9
pg. 
e75961
 
181
Wildhagen
 
KC
García de Frutos
 
P
Reutelingsperger
 
CP
, et al. 
Nonanticoagulant heparin prevents histone-mediated cytotoxicity in vitro and improves survival in sepsis.
Blood
2014
, vol. 
123
 
7
(pg. 
1098
-
1101
)
182
Jain
 
S
Pitoc
 
GA
Holl
 
EK
, et al. 
Nucleic acid scavengers inhibit thrombosis without increasing bleeding.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
32
(pg. 
12938
-
12943
)
183
Matafonov
 
A
Leung
 
PY
Gailani
 
AE
, et al. 
Factor XII inhibition reduces thrombus formation in a primate thrombosis model.
Blood
2014
, vol. 
123
 
11
(pg. 
1739
-
1746
)
Sign in via your Institution