Skip to main content
Research and Practice in Thrombosis and Haemostasis logoLink to Research and Practice in Thrombosis and Haemostasis
. 2018 May 24;2(3):549–557. doi: 10.1002/rth2.12109

The coagulation system in host defense

Silvio Antoniak 1,
PMCID: PMC6046589  PMID: 30046760

Abstract

The blood coagulation system and immune system of higher organisms are thought to have a common ancestral origin. During infections, the blood coagulation system is activated and components of the hemostatic system are directly involved in the immune response and immune system modulations. The current view is that the activation of coagulation is beneficial for infections with bacteria and viruses. It limits pathogen dissemination and supports pathogen killing and tissue repair. On the other hand, over‐activation can lead to thrombosis with subsequent depletion of hemostatic factors and secondary bleeding. This review will summarize the current knowledge on blood coagulation and pathogen infection with focus on most recent studies of the role of the different parts of the blood coagulation system in selected bacterial and viral infections.

Keywords: coagulation, hemostasis, infection, inflammation, peritonitis, pneumonia


Essentials.

  • Blood coagulation system is activated during infections.

  • Components of the coagulation system directly interact with the immune system.

  • Activation of coagulation system limits pathogen dissemination and supports pathogen killing.

  • Overactivation can contribute to infection pathology due to thrombosis and bleeding complications.

1. INTRODUCTION

1.1. Coagulation cascade

Blood coagulation is initiated by exposure of blood to the transmembrane protein tissue factor (TF).1 Under normal conditions, TF is not expressed by cells, such as circulating blood cells and endothelial cells, which are in direct contact with plasma‐circulating coagulation protease zymogens.1, 2 However, subendothelial cells including pericytes, fibroblasts, and smooth muscle cells constitutively express high levels of TF.3 This separation of extravascular TF and circulating plasma‐clotting factors prevents an inappropriate activation of coagulation under basal conditions. In certain tissues perivascular TF is already in complex with the plasma coagulation protease FVII/FVIIa which may enhance coagulation initiation.4 Upon vessel injury, the blood plasma coagulation factors come into contact with the TF:FVIIa complex which converts FX to FXa.5, 6 Importantly, under pathologic conditions TF can also be expressed by monocytes and transferred via membrane‐derived microvesicles (MV) to other cells, including endothelial cells, platelets, and possibly neutrophils.1 The intrinsic pathway consist of FXII, FXI, and FIX with its cofactor FVIII. Historically, it was thought that this pathway is activated by negatively charged artificial and biological surfaces, such as artificial valves and polyphosphates, within the blood independent of vessel injury.7 The intrinsic pathway is now seen as an amplification loop that enhances generation of FXa through the FIXa:FVIIIa tenase complex.1 FXa forms the prothrombinase complex with FVa and mediates the conversion of prothrombin to thrombin. FXa, its cofactor FVa, and thrombin form the common pathway leading to thrombin‐mediated cleavage of fibrinogen to fibrin which is then cross‐linked by activated FXIII forming a stable clot.8

Nowadays, a clear separation of the extrinsic and the intrinsic pathways is difficult since it was shown that the TF:FVIIa complex can also lead to activation of the intrinsic coagulation protease FIX and thrombin can activate FXI.9 FXII does not play a role hemostasis but can contribute to thrombosis as well as inflammatory responses.10 Importantly, there is an ongoing effort to develop new anticoagulants targeting the intrinsic pathway, since deficiencies or inhibition of FXI‐ and FXII‐reduced thrombosis in experimental and clinical studies without a major impact on hemostasis, e.g, spontaneous bleedings.10, 11

1.2. Coagulation‐dependent signaling

Besides leading to a stable clot, active proteases generated during the coagulation cascade can directly induce cell‐specific signaling via the cleavage of protease‐activated receptors (PARs).12 PARs form a receptor family with four members, PAR1‐4.13 Briefly, the TF:FVIIa complex and FXa can activate PAR2.14 Thrombin is the primary activator of PAR1, PAR3, and PAR4. Further, FXa can activate PAR1. PARs are ubiquitously expressed within the body in similar patterns across mammals.13, 14 However, there is a significant difference between human and rodent PAR expression on platelets. Human platelets express PAR1 and PAR4 whereas murine platelets express PAR3 and PAR4.14

Several studies showed that PARs modulate innate immune responses by crosstalk with toll‐like receptors (TLRs).15, 16, 17, 18, 19 Antibacterial TLR4 signaling is enhanced by PAR2, whereas PAR2 reduces antiviral responses via TLR3.15, 16, 19 This PAR2‐dependent TLR modulation is thought to be through direct receptor:receptor interaction.16, 19 In addition, we have shown that coagulation‐dependent PAR1 signaling enhances TLR3‐dependent antiviral responses.17, 18

Unfortunately, experimental studies with global PAR‐deficient mice were not able to reveal any major contribution of PARs to the pathologies of bacterial infection or endotoxemia.20 In contrast to bacterial infections, there are clear phenotypes in PAR1‐ and PAR2‐deficient mice in different viral infection models.14, 15, 16, 17, 18 It could be speculated that host proteases, such as thrombin, stimulate the antiviral response via PAR1, whereas viral proteases may activate PAR2 in an attempt to dampen the antiviral response of the host and increase virus infectivity.17

Newer mouse models with different cleavage‐insensitive PAR1 and PAR2 mutant knock‐in mice might uncover a biased protease‐dependent signaling to certain infections which were hidden in global PAR1 or PAR2 knock‐out mice.21, 22

1.3. Infections and the activation of coagulation

The coagulation system is activated in response to infection by a variety of different pathogens, including bacteria and viruses (Tables 1 and 2).14, 18, 20, 23, 24 This response appears to have developed as a host defense system to limit the spread of the pathogen. During infections, there is an interplay between blood coagulation, immune cells, and platelets to restrict dissemination of pathogens within the body.9, 14, 25 Activation of coagulation coincides with the recruitment of leukocytes where clot components, such as fibrin, serve as a scaffold for adherence and migration of cells. Leukocytes themselves can enhance coagulation by expressing TF and by releasing TF+MV. More importantly, neutrophils release neutrophil extracellular traps (NETs) after activation.9, 26 NETs are composed of nuclear DNA, histones, and several neutrophil enzymes including elastase. NETs were shown to have important antibacterial and potential antiviral functions and, due to their negative charge, also a coagulation‐enhancing activity.9, 26, 27 Thus, leukocytes are thought to be a major player in the cross‐communication between blood coagulation and the immune response.

Table 1.

Role of the blood coagulation system in bacterial infections

Infection Observation References
Bacterial pneumonia
  • Bacterial infection of lung and endotoxemia leads to local activation of coagulation

  • Lung epithelial TF maintains tissue hemostasis after local LPS challenge

  • Local FVIIa administration reduces pulmonary bleeding

  • Myeloid TF does not contribute to activation coagulation in lungs after local LPS challenge

  • Myeloid TF has no role in lung injury after Klebsiella infection

  • Myeloid TF reduces macrophage recruitment into lung after local LPS challenge

  • Myeloid TF reduces lung CXCL1 expression during Klebsiella infection

  • Myeloid TF controls Mycobacterium tuberculosis growth but global TF deficiency (LowTF mice) has no effect

  • FXII−/− mice are protected in Klebsiella pneumonia

  • FXII/FXIIa has no effect on murine and human neutrophil phagocytosis

  • FXII does not contribute to coagulation activation in Klebsiella and Streptococcus pneumoniae

  • FXI deficiency results in higher mortality during Klebsiella and Streptococcus pneumonia associated with higher bacterial out growth and inflammatory response

  • FXI does not contribute to coagulation activation in Klebsiella and Streptococcus pneumonia

  • FXIa is needed for phagocytosis of bacteria by murine and human neutrophils

  • FXI might be activated by thrombin generated via the extrinsic pathway

  • Thrombin inhibition by dabigatran etexilate increased Klebsiella infection but has no effect on activation of coagulation, thrombocytopenia and fibrin deposition

  • Thrombin mediated platelet:neutrophil interaction is needed to limit Klebsiella growth

  • Fibrin degradation/clot lysis due to bacterial proteases leads to increased bleeding tendencies in cystic fibrotic lungs

20, 48, 54, 55, 56, 57, 58, 60, 64
Bacterial peritonitis
  • Myeloid and perivascular TF contributes to systemic activation of coagulation

  • TF contributes to tissue injury and mortality during sepsis

  • TF inhibition mediates survival benefits in endotoxemia

  • FVII consumption causes bleeding and decrease survival

  • FXIIa inhibition does not reduce DIC induced by E. coli infection

  • FXIIa inhibition reduces septic‐induced hypotension and shock

  • CLP leads to FXI‐dependent FXII activation

  • FXI−/− mice have increased survival associated with reduced inflammation in CLP

  • FXI does not contribute to CLP‐mediated DIC

  • Thrombin inhibition does not reduce end‐organ damage in sepsis

  • FV Leiden+/− mice have survival advantage in endotoxemia and sepis caused by Staphlococcus aurens and Yersinia pestis but not CLP and E. coli infection

  • Fibrin deposition limit bacterial dissemination

  • FibAEK mice exhibit reduced S. aureus clearance

  • FibγΔ5 mice exhibit improved survival after S. aureus infection

20, 26, 29, 44, 58, 68, 69, 70, 71, 72, 73, 75, 76, 78
Bacterial Skin Infection
  • FV and fibrinogen deficiency results in increases Streptococcus pyogenes infection

  • FXIII−/− mice exhibit increased Streptococcus pyogenes infection

  • FXIII needed to immobilize bacteria by crosslinking bacterial proteins to fibrinogen/fibrin

  • FXIII mediates innate immune responses to S. pyogenes infection

79, 80, 81

Table 2.

Role of the blood coagulation in viral infections

Infection Observation References
Viral pnemuonia
  • Increased TF expression in the lung

  • Local and systemic activation of coagulation

  • Reduced levels of TF cause increased pulmonary bleeding and mortality in H1N1 IAV infected mice

  • Lung epithelial TF maintains lung hemostasis in H1N1 IAV infected mice

  • Myeloid as well as endothelial/hematopoietic cell TF does not contribute to activation of coagulation in lungs during sub‐lethal H1N1 IAV infection in mice

  • TF+ MV are associated with increased mortality in severe H1N1 IAV infected patients

  • FXII−/− exhibited increased mortality during H1N1 IAV infection

  • FIX deficiency has no effect survival during H1N1 IAV infection

  • Thrombin inhibition with dabigatran etexilate has no effect on survival but reduced local activation of coagulation after H1N1 IAV infection in mice

  • Warfarin increases pulmonary hemorrhages and death in H1N1 IAV infected mice

  • Warfarin increases vascular permeability in H1N1 IAV infected mice lungs

  • Reduced fibrinogen/fibrin levels lead to increased H1N1 IAV infection in mice

23, 39, 82, 83
HIV
  • HIV infection is associated with activation of coagulation

  • HIV leads to TF expression on CD14+ CD16 CCR2+ monocytes

  • Higher cardiovascular risk in controlled HIV infection when D‐dimer is increased

  • On‐going thrombin‐PAR1 signaling on CXCR1+ CD8+ T cells in HIV

  • Thrombin‐PAR1 signaling enhance anti‐viral responses to dsRNA in mice

  • Thrombin increases T cell receptor mediated IFNγ expression on CD8+ T cells

  • Thrombin‐PAR1 signaling increases T Cell motility and cytokine expression

17, 87, 88, 89, 90, 91, 92, 93

Sepsis is a clinical condition as response to an acute bacterial or viral infection in the blood, with ongoing activation of the immune and coagulation system. Unfortunately, overactivation of the coagulation system in acute bacteremia and viremia can lead to disseminated intravascular coagulation (DIC), microvascular thrombosis–induced hypoxia that contributes to multiorgan failure, septic shock, and death.24, 25 Hemorrhages occur due to consumption of coagulation factors and platelets, resulting from ongoing intravascular activation of the hemostatic system.28

Fibrin(ogen) has a central role in hemostasis and thrombosis but it also contributes to multiple physiologic and pathologic processes beyond blood coagulation.29, 30 Reduced fibrin(ogen) levels are a predictor for hemorrhagic complications.31 In addition, fibrin clots were shown to be a strong inducer of a proinflammatory response of in clot‐embedded monocytes and timely degradation via fibrinolysis can dampen this inflammatory response. The reported inflammatory response was thrombin‐independent but fibrin‐dependent.32 Innate immune cells responding via the integrin receptor αMβ2 (CD11b/CD18, Mac‐1) to the γ chain of fibrin(ogen) by phagocytosis, generation of reactive oxygen species and NFκB–mediated gene expression of proinflammatory mediators.33 Interestingly, a report showed that soluble fibrin(ogen) can bind and induce signaling in neutrophils independently of Mac‐1 in vitro which suggests an additional fibrin(ogen) receptor.34 Mac‐1 was further identified as a surface receptor for dsRNA on macrophages mediating TLR3‐dependent and ‐independent immune responses.35 The dsRNA:Mac‐1 interaction was blocked by treatment of cells with fibrin(ogen).35 This observation suggests that high levels of soluble fibrin(ogen) might saturate Mac‐1 and therefore reduce the innate immune response to dsRNA in viral infection. Finally, fibrin degradation products (FDP) are potent chemotactic signals for neutrophils and other leukocytes.36, 37 In addition, FDP are able to enhance as well as inhibit platelet function/aggregation.38

In the past, research was focused on the effect of endotoxemia and bacteremia/sepsis on the coagulation system with regards to DIC, septic shock, and bleeding complications. Newer studies have tried to understand its protective role in viral infections, such as H1N1 influenza A virus (IAV), Ebola virus, and emerging viral pathogens including Dengue and Zika virus.14, 23, 25, 39, 40, 41, 42

Studies showed that vascular TF expression can be induced by pathogen‐associated molecular patterns, including bacterial lipopolysaccharides (LPS) and viral dsRNA which leads to activation of coagulation in vitro and in vivo.43, 44 TF expressed by monocytes/macrophages is the major source of pathologic TF in bacteremia/sepsis and endotoxemia that leads to aberrant coagulation and inflammation.20, 44 The contribution of endothelial cells to activation of coagulation through expression of TF in vivo is controversial.1, 2 In addition, TF is associated with NETs suggesting a direct link between NETs and the extrinsic pathway.1, 45 Neutrophil elastase, which is released during sepsis, can degrade tissue factor pathway inhibitor, the inhibitor of the TF pathway, which may further enhance coagulation.26 Furthermore, case studies reported that FVII consumption and uncontrolled bleeding during sepsis can be reduced and survival improved by systemic administration of additional FVIIa.46, 47 Occurrence of diffuse pulmonary bleeding can be reduced by local administration of FVIIa into the airspace of the lung.48

The role of the intrinsic pathway of coagulation in inflammatory responses was recently summarized in detail by others.10, 49 However, there are only limited data available on the role of the intrinsic coagulation pathway and its members FIX, FXI, and FXII in immune responses to viral infections. Studies proposed that depending on the mode of activation, FXII can either trigger blood coagulation via activation of FXI or activate the kallikrein‐kinin system (KKS). When FXII is bound to an activating surface, the classic activation, FXII is cleaved and activated by plasma prekallikrein/kallikrein in complexing with high molecular weight kininogen (HK) which subsequently leads to FXIa generation.10, 50 In addition, enveloped viruses were shown to enhance intrinsic pathway activation in vitro.51 However, FXII can also be activated by an alternative mechanism via proteases, including elastase and plasmin.50 Certain bacteria were shown to express specific LPS, polyphosphates, elastase, or plasminogen activators to trigger bradykinin production via FXII activation.50, 52, 53 The alternative activation mechanism of FXII results in a significant reduced activation of coagulation and shifting FXIIa towards its pro‐inflammatory role.50

The remainder of this review will focus on the role of the coagulation cascade in infections with selected pathogens with particular attention paid to the intersection of the hemostatic system with antibacterial and antiviral immune responses (Tables 1 and 2).

1.4. Bacterial pneumonia

Pneumonia studies with the Gram‐negative Klebsiella pneumoniae or the Gram‐positive Streptococcus pneumoniae are widely used to investigate local pathogen‐host interactions (Table 1). Both bacteria were shown to lead to local activation of coagulation and fibrin deposition in the lungs.54, 55 Interestingly, while systemic inhibition of TF was beneficial in sepsis and endotoxemia, a global deficiency of TF (LowTF mice) during LPS administration into the lung led to increased pulmonary hemorrhages and lung inflammation.20, 56 Tissue hemostasis and lung‐dependent activation of coagulation during local endotoxemia is mediated by TF expressed on lung epithelial cells and not myeloid cells.57 However, TF deficiency on both cell types did not significantly affect local lung inflammation.57 In line with this, myeloid TF had no effect on Klebsiella pneumonia–induced lung injury.58 Notably, a lack of myeloid TF increased the expression of KC/CXCL1, a neutrophil chemoattractant and the murine homolog to human IL‐8, in the lung after Klebsiella infection and local LPS administration.57, 58 This suggests that myeloid TF is a negative regulator for macrophage infiltration into the alveolus during bacterial infection.57 Moreover, Kral‐Pointner et al reported in acid‐induced lung injury a myeloid TF‐dependent anti‐inflammatory effect within the lung. Mice lacking myeloid cell TF exhibited increased neutrophil accumulation in the lung. Furthermore, in vitro studies showed that myeloid TF dampened NFκB‐dependent responses after hydrochloric acid stimulation.59 Interestingly, in infection, myeloid TF was needed to control bacterial growth.60 A lack of myeloid cell TF resulted in less fibrin deposition and a M2 macrophage phenotype within the lung.60 However, when using a global TF deficient mouse (LowTF mice) this difference was not detectable which could be due a low but still sufficient TF expression in macrophages.61 Further, Rauch's group found that the FXa inhibitor fondaparinux increased a M2 macrophage phenotype in the mouse heart during viral myocarditis.62 This suggest that myeloid expressed TF suppresses the differentiation into an anti‐inflammatory M2 macrophage. It is not clear if this TF:FXa effect is PAR‐dependent.

Stroo et al used FXI and FXII deficient mice to investigate the role of both factors in Klebsiella and Streptococcus pneumonia and found that a lack of FXI resulted in higher mortality and enhanced bacterial outgrowth in both models.55 This observation was accompanied by increased inflammatory responses in FXI−/− mice.55 In contrast to the findings with FXI−/− mice, FXII−/− mice were protected only in Klebsiella pneumonia associated with improved survival and reduced bacterial burden. Both models showed that a deficiency of either FXI or FXII did not reduced the local activation of coagulation. Furthermore, the authors reported that active FXI was needed for phagocytosis of bacteria by murine and human neutrophils, whereas FXII or FXIIa‐dependent FXI activation had no effect on phagocytosis.55

It was shown that thrombin activity leads to endothelial cell activation in Klebsiella infection.54 Interestingly, thrombin inhibition and fibrin depletion resulted in increased Klebsiella infection in mice associated with increased bacterial outgrowth and dissemination leading to higher mortality.54 However, the thrombin inhibitor dabigatran had no effect on neutrophil recruitment, activation, and NET formation, but it dampened coagulation activation measured by reduced D‐dimer and thrombin‐antithrombin (TAT) levels, and fibrin depositions within the lung.54 Whole blood assays showed that the combination of active thrombin, platelets, and neutrophils were essential to limit Klebsiella growth.54 Interestingly, the authors observed that thrombin‐mediated PAR1 activation on platelets reduced Klebsiella growth in human blood by enhancing platelet‐neutrophil interaction.54 Further, thrombin inhibition reduced platelet‐neutrophil interaction in Klebsiella pneumonia but did not effected thrombocytopenia.54

These findings suggested that extrinsic pathway generated thrombin mediates fibrin polymerization and platelet‐neutrophil interactions are essential for protective immune responses in at least Klebsiella pneumonia–derived sepsis.54 Furthermore, FXI is activated by extrinsic pathway generated thrombin independently of FXIIa in bacterial pneumonia which influences the antibacterial function of neutrophils.55

Importantly, bacteria can support the activation of plasminogen on their surface.63 Increased clot lysis and fibrin degradation would lead to bacterial dissemination. Cystic fibrosis is associated with specific bacterial colonization of the lung and can cause hemoptysis a hallmark of “cepacia syndrome”.64 Within the cystic fibrosis lung, bacterial proteases as well as neutrophil elastase are generated/released which were shown to degrade fibrin.64 Cystic fibrosis hemoptysis is therefore a result of infection‐driven immune responses causing a failure of lung integrity and affecting lung hemostasis, which subsequently leads to hemorrhages into the airway lumen during the phase of acute infection.64, 65, 66

1.5. Bacterial peritonitis

To analyze the effect of systemic bacteremia/sepsis and endoxemia, mice are subjected to intraperitoneal or intravenous injection of bacteria or endotoxin as well as cecal ligation and puncture (CLP) surgery. The majority of the studies that have analyzed the role of the coagulation system in peritonitis and sepsis use Staphylococcus aureus, Yersinia pestis and Escherichia coli (Table 1). The interaction between the host's coagulation/immune system and S. aureus were extensively reviewed recently in detail.67 It was repeatedly shown that the TF pathway is essential for the induction of coagulation in endotoxemia and sepsis, which subsequently leads to tissue injury and mortality.20, 44, 68, 69 Further, inhibition of TF‐dependent activation was reported to improve survival in endotoxemia and sepsis.20 The major contributor in endotoxemia induced systemic coagulation activation is TF expressed by myeloid cells as well as by cells of unknown perivascular origin.44 Interestingly, Bastarache's group was not able to show that myeloid TF had any role in indirect lung injury during endotoxemia and CLP sepsis.58 In addition, the prothrombotic phenotype of FV Leiden heterozygosity in mice poses a survival advantage in endotoxemia and sepsis caused by S. aureus and Y. pestis but not by CLP or E. coli.70, 71 The authors proposed that FV Leiden has anti‐fibrinolytic effects which opposes the bacterial fibrinolytic virulence factors.70 Interestingly, endothelial PC receptor deficiency but not PAR1 deficiency abrogated the survival advantage of heterozygous FV Leiden mice.70

It seems that the intrinsic pathway does not play any significant role in the activation of coagulation but contributes to inflammation during sepsis. For instance, an anti‐FXIIa antibody or FXI deficiency did not block E. coli or CLP‐induced DIC, respectively.72, 73 Only one study showed that inhibition of FXIIa‐dependent FXI activation via the inhibitory antibody 14E11 reduced thrombin generation, platelet consumption, cytokine expression and resulted in improved survival of CLP mice.74 Furthermore, FXIIa inhibition reduced sepsis‐induced hypotension and shock.50 Also, FXIa was shown to induce cytokine responses after CLP in mice.73, 74 Thus, FXI−/− mice exhibited increased survival with reduced CLP‐induced cytokine expression compared to WT mice. Furthermore, FXIa can activate FXII leading to enhanced activation of the intrinsic pathway and KKS.73 Indeed, CLP caused a reduction in FXII and prekallikrein plasma levels in WT mice but not FXI−/− mice.73 These data suggest that FXI or FXII inhibition might be beneficial by reducing inflammatory responses in polymicrobial abdominal sepsis but not in bacterial infection of the lung.55, 73, 74

Studies showed an important role for fibrin(ogen) and FXIII to limit bacterial dissemination (Table 1).26, 29, 75, 76 To analyze the role of fibrin in bacterial infections, Prasad and colleagues generated mice carrying a thrombin‐cleavage resistant fibrinogen Aα chain (FibAEK). While these mice have normal levels of circulating fibrinogen levels and support normal platelet:fibrinogen interaction they are unable to produce fibrin polymers.77 Interestingly, FibAEK mice exhibited a profound impediment in S. aureus clearance following intraperitoneal infection similar to Fib−/− mice but had a significant infection dose‐dependent survival advantage over Fib−/− mice following peritonitis challenge.77 This indicates that the fibrin polymerization is critical for the antibacterial action while circulating fibrinogen has additional protective functions possible due to platelet interaction. In addition, mice lacking the last five amino acids of the fibrinogen γ chain (FibγΔ5) exhibited improved survival after S. aureus infection compared to WT and Fibγ390‐396A.78 Platelet and platelet integrin receptor subunit αIIb deficient mice established that the survival benefits observed in FibγΔ5 mice were largely independent of platelet αIIbβ3‐mediated engagement of fibrinogen.

1.6. Bacterial skin infection

Skin infection with Streptococcus pyogenes (Group A streptococcus) is a major public health concern. While local infection is mostly uncomplicated, a systemic dissemination is associated with streptococcal toxic shock syndrome. Ginsburg's group reported that FV and fibrinogen deficiency in mice resulted in increased S. pyogenes infection, suggesting that FV‐dependent fibrin deposition was needed to reduce pathogen dissemination (Table 1).79 However, FV Leiden had no effect on S. pyogenes infection.79

In line to the findings with fibrinogen deficiency, FXIII deficiency during S. pyogenes infection evokes a pathologic inflammatory reaction causing massive neutrophil influx at the side of infection.80 In addition, FXIII is essential for immobilization of bacteria, such as S. pyogenes, within the fibrin network which prevents bacterial dissemination and reduced inflammatory overreaction.81 Furthermore, local FXIII application at the site of infection resulted in a reduction bacterial dissemination indicating that FXIII mediates protection during early S. pyogenes skin infection by supporting early innate immune response.81

There are compelling data that reducing coagulation activation can improve the outcome in endotoxemia (Table 1). On the other hand, during bacterial sepsis the activation of coagulation and local thrombosis/fibrin deposition can improve host survival by limiting dissemination of certain bacteria species, including K. pneumoniae, S. pneumoniae, S. pyogenes and E. coli. In general, fibrin generation initiated by the host leads to bacterial entrapment reducing bacterial dissemination and increasing pathogen killing by leukocytes. Only a few bacterial pathogens, such as S. aureus, developed virulence factors to initiate fibrin generation for evade immune system recognition.67

1.7. Viral pneumonia

Recently, we showed that TF is induced in the lung after H1N1 IAV infection in mice which led to increased activation of coagulation via increased TF activity in the lung and by TF+MV in the bronchoalveolar lavage fluid (BALF) (Table 2).23 The induction of TF and activation of coagulation was abolished in mice with a global TF deficiency (LowTF mice) and mice with a TF deletion in lung epithelial cells suggesting that lung epithelial cells are the main source for inducible TF in the lung and driver of coagulation after H1N1 IAV infection.23 Furthermore, LowTF mice and mice lacking lung epithelial cell TF presented with increased alveolar hemorrhages and death in sub‐lethal H1N1 IAV infection.23 Importantly, we could not find any contribution for myeloid, endothelial, or hematopoietic TF on the activation of coagulation nor survival after H1N1 IAV infection.23 The current studies indicating that H1N1 IAV infection is a hemostatic challenge and epithelial cell TF‐dependent fibrin deposition mediates lung hemostasis.23, 39 Indeed, reduction of fibrin(ogen) levels with the snake venom ancrod led to an increase of H1N1 IAV infection pathology in mice.82 However, during severe H1N1 IAV infection, increased TF levels, possible on macrophages, can be deleterious due to increased TF activity on MV in plasma which was associated with increased mortality in H1N1 IAV infected patients.83

We found that dabigatran decreased activation of coagulation in the lung measured by TAT levels in the BALF of H1N1 IAV infected mice which was associated with increased pulmonary hemorrhages.39 However, we could not find any differences in the survival between dabigatran and placebo treated H1N1 IAV infected mice.39 Importantly, we observed that anticoagulation with the vitamin‐K antagonist warfarin increased pulmonary hemorrhages and mortality of H1N1 IAV infected mice.39 Anticoagulation with warfarin unselective reduces the vitamin K–dependent procoagulant factors prothrombin, FVII, FIX, FX, and the anticoagulant factors protein C and S, whereas dabigatran only inhibits thrombin activity. APC and FVII were shown to facilitate vascular protection via the endothelial protein C receptor and PAR1 in certain disease models. We found that warfarin but not dabigatran significantly increased vascular permeability in the lung after H1N1 IAV infection compared to H1N1 IAV alone.39 In support of our findings, APC administration reduced Dengue virus mediated endothelial permeability and inflammatory response,84 which suggests that APC may be protective during Dengue virus and possible other viral infections. However, the action of APC might be more complicated since inhibition of APC was shown to worsen lung histopathology but lowered neutrophil influx and delayed mortality during lethal IAV infection in mice.85 We found that PAR1−/− mice exhibited increased inflammation in the lung early after H1N1 IAV infection.18 However, thrombin inhibition had no effect on inflammation and survival after H1N1 IAV infection suggesting that thrombin might not be the major PAR1 activator in this model.39 As mentioned before, cleavage‐resistant PAR1 knock‐in mice might reveal the relative contribution of thrombin‐ vs APC‐dependent PAR1 activation in H1N1 IAV infections.21

With regard to the intrinsic pathway in H1N1 IAV infection, we reported that FIX deficiency had no effect on survival after infection.23 Interestingly, we observed that lack of FXII in mice increased the mortality after sub‐lethal H1N1 IAV infection (Tatsumi et al, unpublished data). This discrepancy suggests that the intrinsic part of the blood coagulation was not needed to contribute to hemostasis‐dependent survival after H1N1 IAV infection.23 However, it is possible that FXIIa‐dependent activation of the KKS mediates protection in H1N1 IAV infection. Surprisingly, we were not able to see any differences in the survival between HK deficient and WT control mice suggesting that FXII‐dependent KKS activation is not needed for a positive outcome after H1N1 IAV infection (Tatsumi et al, unpublished data). There might be important FXII function independent of FXI and the KKS which could explain the higher mortality in FXII−/− mice in H1N1 IAV infection. For instance, neutrophils were shown to express FXII and that the FXII zymogen acts via uPAR as modulator of neutrophil adhesion and chemotaxis.86 Unfortunately, the lack of in vivo studies makes it difficult to definitely conclude if the intrinsic pathway has any significant role in viral infections.

1.8. HIV

HIV infection is associated increased TAT and D‐dimer levels in plasma suggesting an ongoing activation of coagulation in HIV infection (Table 2).87, 88, 89 Further, the coagulation activation marker correlated with virus load and monocyte TF expression in HIV‐infected patients.90 Interestingly, TF expression seems to be restricted to CD14+ CD16 CCR2+ monocytes.91 Thrombin mediates the crosstalk between the coagulation system and the adaptive immune system at sites of vascular injury via PAR1 increasing T cell motility and proinflammatory cytokine production.92 Importantly, HIV‐infected antiretroviral therapy recipients exhibit, even with suppressed viremia, increased risk for cardiovascular disease due to ongoing thrombin‐mediated signaling through PAR1 on CXCR1+ CD8+ T cells.93 Thrombin further directly enhanced T cell receptor–mediated interferon (IFN) γ production by purified CD8+ T cells.93

Currently, there are two ongoing clinical studies with FXa inhibitor edoxaban (TACTICAL‐HIV, NCT02339415) or the PAR1 inhibitor vorapaxar (ADVICE, NCT02394730) in patients with HIV infection who are successfully treated with combination antiretroviral therapy. Both studies will compare the safety and efficacy of either edoxaban or vorapaxar in reducing markers of immune system activation in HIV disease. In support of these studies, we showed that thrombin inhibition with dabigatran or PAR1 deficiency reduced the innate immune responses in virus‐like stimulation with a dsRNA mimetic in mice.17

2. CONCLUSION

In the past, studies have tried to understand whether the activation of coagulation during infections was merely a side effect contributing to infection pathology or if it is an active part in the host's immune response to the pathogen. Nowadays, it is accepted that there is a continuous crosstalk between the immune system with blood coagulation components. Both systems are closely intertwined and essential for an effective immune response to limit the infection. However, as with all fine‐tuned systems, once out of control, overactivation can outweigh the beneficial effects by inducing thrombotic complication, excessive inflammation, and tissue damage. Further studies should investigate how to avoid the coagulation overactivation without compromising the beneficial contribution. With regard to the potential clinical significance, it is possible that interference with the PAR1 pathway by direct thrombin inhibitors or PAR1 inhibitors may increase the risk and severity of viral infection, whereas FXa or PAR2 pathway inhibition could mediate a beneficial outcome.14, 62 However, the importance of the PAR1 and PAR2 signaling pathway might be only applicable for an initial viral infection or in immunocompromised individuals. Importantly, there are conflicting data about if the FXa inhibitor rivaroxaban increases the occurrence of early periprosthetic joint infections.94, 95 A recent meta‐analysis for the risk of infection with new oral anticoagulants did not to find any association.96 However, the authors stressed that their analysis was limited due to the potential selective reporting bias in the analyzed studies.96 Further large prospective studies should properly address if the new oral anticoagulants increase the risk for certain bacterial or viral infections.

RELATIONSHIP DISCLOSURE

The author has nothing to disclose.

ACKNOWLEDGMENTS

Special thanks to Dr. S. Grover and Dr. N. Mackman for critical reading of the manuscript. The study was supported by grants of the American Heart Association 14BGIA20380134 and the National Institutes of Health R01 HL142799‐01. There are no financial interests.

Antoniak S. The coagulation system in host defense. Res Pract Thromb Haemost. 2018;2:549–557. 10.1002/rth2.12109

REFERENCES

  • 1. Grover SP, Mackman N. Tissue factor: an essential mediator of hemostasis and trigger of thrombosis. Arterioscler Thromb Vasc Biol. 2018;38:709–25. [DOI] [PubMed] [Google Scholar]
  • 2. Antoniak S, Mackman N. Editorial commentary: tissue factor expression by the endothelium: coagulation or inflammation? Trends Cardiovasc Med. 2016;26:304–5. [DOI] [PubMed] [Google Scholar]
  • 3. Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of tissue factor in human tissues. Implications for disorders of hemostasis and thrombosis. Am J Pathol. 1989;134:1087–97. [PMC free article] [PubMed] [Google Scholar]
  • 4. Hoffman M, Colina CM, McDonald AG, Arepally GM, Pedersen L, Monroe DM. Tissue factor around dermal vessels has bound factor VII in the absence of injury. J Thromb Haemost. 2007;5:1403–8. [DOI] [PubMed] [Google Scholar]
  • 5. Josso F, Prou‐Wartelle O. Interaction of tissue factor and factor VII at the earliest phase of coagulation. Thromb Diath Haemorrh Suppl. 1965;17:35–44. [PubMed] [Google Scholar]
  • 6. Lu G, Broze GJ Jr, Krishnaswamy S. Formation of factors IXa and Xa by the extrinsic pathway: differential regulation by tissue factor pathway inhibitor and antithrombin III. J Biol Chem. 2004;279:17241–9. [DOI] [PubMed] [Google Scholar]
  • 7. Maas C, Oschatz C, Renne T. The plasma contact system 2.0. Semin Thromb Hemost. 2011;37:375–81. [DOI] [PubMed] [Google Scholar]
  • 8. Walton BL, Byrnes JR, Wolberg AS. Fibrinogen, red blood cells, and factor XIII in venous thrombosis. J Thromb Haemost. 2015;13(Suppl 1):S208–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gaertner F, Massberg S. Blood coagulation in immunothrombosis‐At the frontline of intravascular immunity. Semin Immunol. 2016;28:561–9. [DOI] [PubMed] [Google Scholar]
  • 10. Maas C, Renne T. Coagulation factor XII in thrombosis and inflammation. Blood. 2018;131(17):1903–9. [DOI] [PubMed] [Google Scholar]
  • 11. Fredenburgh JC, Gross PL, Weitz JI. Emerging anticoagulant strategies. Blood. 2017;129:147–54. [DOI] [PubMed] [Google Scholar]
  • 12. Coughlin SR. Thrombin signalling and protease‐activated receptors. Nature. 2000;407:258–64. [DOI] [PubMed] [Google Scholar]
  • 13. Antoniak S, Sparkenbaugh E, Pawlinski R. Tissue factor, protease activated receptors and pathologic heart remodelling. Thromb Haemost. 2014;112:893–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Antoniak S, Mackman N. Multiple roles of the coagulation protease cascade during virus infection. Blood. 2014;123:2605–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Nhu QM, Shirey K, Teijaro JR, et al. Novel signaling interactions between proteinase‐activated receptor 2 and Toll‐like receptors in vitro and in vivo. Mucosal Immunol. 2010;3:29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Weithauser A, Bobbert P, Antoniak S, et al. Protease‐activated receptor 2 regulates the innate immune response to viral infection in a CVB3‐induced myocarditis. J Am Coll Cardiol. 2013;62:1737–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Antoniak S, Tatsumi K, Bode M, Vanja S, Williams JC, Mackman N. Protease‐activated receptor 1 enhances poly I: C induction of the antiviral response in macrophages and mice. J Innate Immun. 2017;9:181–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Antoniak S, Owens AP 3rd, Baunacke M, et al. PAR‐1 contributes to the innate immune response during viral infection. J Clin Invest. 2013;123:1310–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Rallabhandi P, Nhu QM, Toshchakov VY, et al. Analysis of proteinase‐activated receptor 2 and TLR4 signal transduction: a novel paradigm for receptor cooperativity. J Biol Chem. 2008;283:24314–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Pawlinski R, Mackman N. Tissue factor, coagulation proteases, and protease‐activated receptors in endotoxemia and sepsis. Crit Care Med. 2004;32:S293–7. [DOI] [PubMed] [Google Scholar]
  • 21. Sinha RK, Wang Y, Zhao Z, et al. PAR1 biased signaling is required for activated protein C in vivo benefits in sepsis and stroke. Blood. 2018;131:1163–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Liang HP, Kerschen EJ, Hernandez I, et al. EPCR‐dependent PAR2 activation by the blood coagulation initiation complex regulates LPS‐triggered interferon responses in mice. Blood. 2015;125:2845–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Antoniak S, Tatsumi K, Hisada Y, et al. Tissue factor deficiency increases alveolar hemorrhage and death in influenza A virus‐infected mice. J Thromb Haemost. 2016;14:1238–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Levi M, van der Poll T. Coagulation and sepsis. Thromb Res. 2017;149:38–44. [DOI] [PubMed] [Google Scholar]
  • 25. Goeijenbier M, van Wissen M, van de Weg C, et al. Review: viral infections and mechanisms of thrombosis and bleeding. J Med Virol. 2012;84:1680–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Massberg S, Grahl L, von Bruehl ML, et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. 2010;16:887–96. [DOI] [PubMed] [Google Scholar]
  • 27. Agraz‐Cibrian JM, Giraldo DM, Mary FM, Urcuqui‐Inchima S. Understanding the molecular mechanisms of NETs and their role in antiviral innate immunity. Virus Res. 2017;228:124–33. [DOI] [PubMed] [Google Scholar]
  • 28. Boral BM, Williams DJ, Boral LI. Disseminated intravascular coagulation. Am J Clin Pathol. 2016;146:670–80. [DOI] [PubMed] [Google Scholar]
  • 29. Ko YP, Flick MJ. Fibrinogen is at the interface of host defense and pathogen virulence in Staphylococcus aureus infection. Semin Thromb Hemost. 2016;42:408–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Pillay J, Kamp VM, Pennings M, et al. Acute‐phase concentrations of soluble fibrinogen inhibit neutrophil adhesion under flow conditions in vitro through interactions with ICAM‐1 and MAC‐1 (CD11b/CD18). J Thromb Haemost. 2013;11:1172–82. [DOI] [PubMed] [Google Scholar]
  • 31. Lord ST. Fibrinogen and fibrin: scaffold proteins in hemostasis. Curr Opin Hematol. 2007;14:236–41. [DOI] [PubMed] [Google Scholar]
  • 32. Campbell RA, Vieira‐de‐Abreu A, Rowley JW, et al. Clots are potent triggers of inflammatory cell gene expression: indications for timely fibrinolysis. Arterioscler Thromb Vasc Biol. 2017;37:1819–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Flick MJ, Du X, Degen JL. Fibrin(ogen)‐alpha M beta 2 interactions regulate leukocyte function and innate immunity in vivo. Exp Biol Med (Maywood). 2004;229:1105–10. [DOI] [PubMed] [Google Scholar]
  • 34. de Almeida VV, Calado A, Rosario HS, Saldanha C. Differential effect of soluble fibrinogen as a neutrophil activator. Microvasc Res. 2012;83:332–6. [DOI] [PubMed] [Google Scholar]
  • 35. Zhou H, Liao J, Aloor J, et al. CD11b/CD18 (Mac‐1) is a novel surface receptor for extracellular double‐stranded RNA to mediate cellular inflammatory responses. J Immunol. 2013;190:115–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Leavell KJ, Peterson MW, Gross TJ. The role of fibrin degradation products in neutrophil recruitment to the lung. Am J Respir Cell Mol Biol. 1996;14:53–60. [DOI] [PubMed] [Google Scholar]
  • 37. Skogen WF, Senior RM, Griffin GL, Wilner GD. Fibrinogen‐derived peptide B beta 1‐42 is a multidomained neutrophil chemoattractant. Blood. 1988;71:1475–9. [PubMed] [Google Scholar]
  • 38. Wilson PA, McNicol GP, Douglas AS. Effect of fibrinogen degradation products on platelet aggregation. J Clin Pathol. 1968;21:147–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Tatsumi K, Antoniak S, Subramaniam S, et al. Anticoagulation increases alveolar hemorrhage in mice infected with influenza A. Physiol Rep. 2016;4(24):e13071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Boyer Chammard T, Schepers K, Breurec S, et al. Severe thrombocytopenia after Zika virus infection, Guadeloupe, 2016. Emerg Infect Dis. 2017;23:696–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Basler CF. Molecular pathogenesis of viral hemorrhagic fever. Semin Immunopathol. 2017;39:551–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Geisbert TW, Hensley LE, Jahrling PB, et al. Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet. 2003;362:1953–8. [DOI] [PubMed] [Google Scholar]
  • 43. Shibamiya A, Hersemeyer K, Schmidt Woll T, et al. A key role for Toll‐like receptor‐3 in disrupting the hemostasis balance on endothelial cells. Blood. 2009;113:714–22. [DOI] [PubMed] [Google Scholar]
  • 44. Pawlinski R, Wang JG, Owens AP 3rd, et al. Hematopoietic and nonhematopoietic cell tissue factor activates the coagulation cascade in endotoxemic mice. Blood. 2010;116:806–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. 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;36:1405–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Martinez J, Cid AR, de la Rubia J, Gimeno R. Treatment of intra‐abdominal bleeding with recombinant activated factor VII in a patient with disseminated intravascular coagulation secondary to septic shock. Blood Coagul Fibrinolysis. 2005;16:297–9. [DOI] [PubMed] [Google Scholar]
  • 47. Schmid S, Friesenecker B, Lorenz I, et al. Administration of recombinant activated factor VII (NovoSeven) in three cases of uncontrolled bleeding caused by disseminated intravascular coagulopathy. Clin Appl Thromb Hemost. 2007;13:313–7. [DOI] [PubMed] [Google Scholar]
  • 48. Heslet L, Nielsen JD, Nepper‐Christensen S. Local pulmonary administration of factor VIIa (rFVIIa) in diffuse alveolar hemorrhage (DAH)—a review of a new treatment paradigm. Biologics. 2012;6:37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Wu Y. Contact pathway of coagulation and inflammation. Thromb J. 2015;13:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Jukema BN, de Maat S, Maas C. Processing of factor XII during inflammatory reactions. Front Med (Lausanne). 2016;3:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Gershom ES, Sutherland MR, Lollar P, Pryzdial EL. Involvement of the contact phase and intrinsic pathway in herpes simplex virus‐initiated plasma coagulation. J Thromb Haemost. 2010;8:1037–43. [DOI] [PubMed] [Google Scholar]
  • 52. Nitzsche R, Rosenheinrich M, Kreikemeyer B, Oehmcke‐Hecht S. Streptococcus pyogenes triggers activation of the human contact system by streptokinase. Infect Immun. 2015;83:3035–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Khan MM, Yamamoto T, Araki H, et al. Pseudomonal elastase injection causes low vascular resistant shock in guinea pigs. Biochim Biophys Acta. 1993;1182:83–93. [DOI] [PubMed] [Google Scholar]
  • 54. Claushuis TA, de Stoppelaar SF, Stroo I, et al. Thrombin contributes to protective immunity in pneumonia‐derived sepsis via fibrin polymerization and platelet‐neutrophil interactions. J Thromb Haemost. 2017;15:744–57. [DOI] [PubMed] [Google Scholar]
  • 55. Stroo I, Zeerleder S, Ding C, et al. Coagulation factor XI improves host defence during murine pneumonia‐derived sepsis independent of factor XII activation. Thromb Haemost. 2017;117:1601–14. [DOI] [PubMed] [Google Scholar]
  • 56. Bastarache JA, Sebag SC, Clune JK, et al. Low levels of tissue factor lead to alveolar haemorrhage, potentiating murine acute lung injury and oxidative stress. Thorax. 2012;67:1032–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Shaver CM, Grove BS, Putz ND, et al. Regulation of alveolar procoagulant activity and permeability in direct acute lung injury by lung epithelial tissue factor. Am J Respir Cell Mol Biol. 2015;53:719–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Shaver CM, Grove BS, Clune JK, Mackman N, Ware LB, Bastarache JA. Myeloid tissue factor does not modulate lung inflammation or permeability during experimental acute lung injury. Sci Rep. 2016;6:22249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Kral‐Pointner JB, Schrottmaier WC, Horvath V, et al. Myeloid but not epithelial tissue factor exerts protective anti‐inflammatory effects in acid aspiration‐induced acute lung injury. J Thromb Haemost. 2017;15:1625–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Venkatasubramanian S, Tripathi D, Tucker T, et al. Tissue factor expression by myeloid cells contributes to protective immune response against Mycobacterium tuberculosis infection. Eur J Immunol. 2016;46:464–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kothari H, Keshava S, Vatsyayan R, Mackman N, Rao LV, Pendurthi UR. Role of tissue factor in Mycobacterium tuberculosis‐induced inflammation and disease pathogenesis. PLoS ONE. 2014;9:e114141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Malz R, Weithauser A, Tschope C, Schultheiss HP, Rauch U. Inhibition of coagulation factor Xa improves myocardial function during CVB3‐induced myocarditis. Cardiovasc Ther. 2014;32:113–9. [DOI] [PubMed] [Google Scholar]
  • 63. Tapper H, Herwald H. Modulation of hemostatic mechanisms in bacterial infectious diseases. Blood. 2000;96:2329–37. [PubMed] [Google Scholar]
  • 64. Reihill JA, Moreland M, Jarvis GE, et al. Bacterial proteases and haemostasis dysregulation in the CF lung. J Cyst Fibros. 2017;16:49–57. [DOI] [PubMed] [Google Scholar]
  • 65. Flume PA, Yankaskas JR, Ebeling M, Hulsey T, Clark LL. Massive hemoptysis in cystic fibrosis. Chest. 2005;128:729–38. [DOI] [PubMed] [Google Scholar]
  • 66. Efrati O, Harash O, Rivlin J, et al. Hemoptysis in Israeli CF patients–prevalence, treatment, and clinical characteristics. J Cyst Fibros. 2008;7:301–6. [DOI] [PubMed] [Google Scholar]
  • 67. Liesenborghs L, Verhamme P, Vanassche T. Staphylococcus aureus, master manipulator of the human hemostatic system. J Thromb Haemost. 2018;16:441–54. [DOI] [PubMed] [Google Scholar]
  • 68. Carraway MS, Welty‐Wolf KE, Miller DL, et al. Blockade of tissue factor: treatment for organ injury in established sepsis. Am J Respir Crit Care Med. 2003;167:1200–9. [DOI] [PubMed] [Google Scholar]
  • 69. Creasey AA, Chang AC, Feigen L, Wun TC, Taylor FB Jr, Hinshaw LB. Tissue factor pathway inhibitor reduces mortality from Escherichia coli septic shock. J Clin Invest. 1993;91:2850–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Kerschen E, Hernandez I, Zogg M, Maas M, Weiler H. Survival advantage of heterozygous factor V Leiden carriers in murine sepsis. J Thromb Haemost. 2015;13:1073–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Kerlin BA, Yan SB, Isermann BH, et al. Survival advantage associated with heterozygous factor V Leiden mutation in patients with severe sepsis and in mouse endotoxemia. Blood. 2003;102:3085–92. [DOI] [PubMed] [Google Scholar]
  • 72. Pixley RA, De La Cadena R, Page JD, et al. The contact system contributes to hypotension but not disseminated intravascular coagulation in lethal bacteremia. In vivo use of a monoclonal anti‐factor XII antibody to block contact activation in baboons. J Clin Invest. 1993;91:61–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Bane CE Jr, Ivanov I, Matafonov A, et al. Factor XI deficiency alters the cytokine response and activation of contact proteases during polymicrobial sepsis in mice. PLoS ONE. 2016;11:e0152968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Tucker EI, Verbout NG, Leung PY, et al. Inhibition of factor XI activation attenuates inflammation and coagulopathy while improving the survival of mouse polymicrobial sepsis. Blood. 2012;119:4762–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Bhattacharya S, Ploplis VA, Castellino FJ. Bacterial plasminogen receptors utilize host plasminogen system for effective invasion and dissemination. J Biomed Biotechnol. 2012;2012:482096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Wang Z, Wilhelmsson C, Hyrsl P, et al. Pathogen entrapment by transglutaminase—a conserved early innate immune mechanism. PLoS Pathog. 2010;6:e1000763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Prasad JM, Gorkun OV, Raghu H, et al. Mice expressing a mutant form of fibrinogen that cannot support fibrin formation exhibit compromised antimicrobial host defense. Blood. 2015;126:2047–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Flick MJ, Du X, Prasad JM, et al. Genetic elimination of the binding motif on fibrinogen for the S. aureus virulence factor ClfA improves host survival in septicemia. Blood. 2013;121:1783–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Sun H, Wang X, Degen JL, Ginsburg D. Reduced thrombin generation increases host susceptibility to group A streptococcal infection. Blood. 2009;113:1358–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Loof TG, Morgelin M, Johansson L, et al. Coagulation, an ancestral serine protease cascade, exerts a novel function in early immune defense. Blood. 2011;118:2589–98. [DOI] [PubMed] [Google Scholar]
  • 81. Deicke C, Chakrakodi B, Pils MC, et al. Local activation of coagulation factor XIII reduces systemic complications and improves the survival of mice after Streptococcus pyogenes M1 skin infection. Int J Med Microbiol. 2016;306:572–9. [DOI] [PubMed] [Google Scholar]
  • 82. Berri F, Rimmelzwaan GF, Hanss M, et al. Plasminogen controls inflammation and pathogenesis of influenza virus infections via fibrinolysis. PLoS Pathog. 2013;9:e1003229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Rondina MT, Tatsumi K, Bastarache JA, Mackman N. Microvesicle tissue factor activity and interleukin‐8 levels are associated with mortality in patients with influenza A/H1N1 infection. Crit Care Med. 2016;44:e574–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Cabello‐Gutierrez C, Manjarrez‐Zavala ME, Huerta‐Zepeda A, et al. Modification of the cytoprotective protein C pathway during Dengue virus infection of human endothelial vascular cells. Thromb Haemost. 2009;101:916–28. [PubMed] [Google Scholar]
  • 85. Schouten M, de Boer JD, van der Sluijs KF, et al. Impact of endogenous protein C on pulmonary coagulation and injury during lethal H1N1 influenza in mice. Am J Respir Cell Mol Biol. 2011;45:789–94. [DOI] [PubMed] [Google Scholar]
  • 86. Stavrou EX, Fang C, Bane KL, et al. Factor XII and uPAR upregulate neutrophil functions to influence wound healing. J Clin Invest. 2018;128:944–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Younas M, Psomas C, Reynes J, Corbeau P. Immune activation in the course of HIV‐1 infection: causes, phenotypes and persistence under therapy. HIV Med. 2016;17:89–105. [DOI] [PubMed] [Google Scholar]
  • 88. Tenorio AR, Zheng Y, Bosch RJ, et al. Soluble markers of inflammation and coagulation but not T‐cell activation predict non‐AIDS‐defining morbid events during suppressive antiretroviral treatment. J Infect Dis. 2014;210:1248–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Funderburg NT, Lederman MM . Coagulation and morbidity in treated HIV infection. Thromb Res. 2014;133(Suppl 1):S21–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Funderburg NT, Mayne E, Sieg SF, et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood. 2010;115:161–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Schechter ME, Andrade BB, He T, et al. Inflammatory monocytes expressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med. 2017;. 10.1126/scitranslmed.aam5441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Hurley A, Smith M, Karpova T, et al. Enhanced effector function of CD8(+) T cells from healthy controls and HIV‐infected patients occurs through thrombin activation of protease‐activated receptor 1. J Infect Dis. 2013;207:638–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Mudd JC, Panigrahi S, Kyi B, et al. Inflammatory function of CX3CR1+ CD8+ T cells in treated HIV infection is modulated by platelet interactions. J Infect Dis. 2016;214:1808–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Brimmo O, Glenn M, Klika AK, Murray TG, Molloy RM, Higuera CA. Rivaroxaban use for thrombosis prophylaxis is associated with early periprosthetic joint infection. J Arthroplasty. 2016;31:1295–8. [DOI] [PubMed] [Google Scholar]
  • 95. Di Benedetto P, Zangari A, De Franceschi D, et al. Rivaroxaban and early periprostethic joint infection: our experience. Acta Biomed. 2017;88:38–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Caldeira D, Costa J, Pinto FJ, Ferreira JJ. The risk of infection with new oral anticoagulants: a meta‐analysis. Int J Cardiol. 2014;172:267–8. [DOI] [PubMed] [Google Scholar]

Articles from Research and Practice in Thrombosis and Haemostasis are provided here courtesy of Elsevier

RESOURCES