Sie sind auf Seite 1von 19

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/323017833

Tissue Factor: An Essential Mediator of Hemostasis and Trigger of


Thrombosis

Article  in  Arteriosclerosis Thrombosis and Vascular Biology · February 2018


DOI: 10.1161/ATVBAHA.117.309846

CITATIONS READS

9 220

2 authors, including:

Steven Philip Grover


University of North Carolina at Chapel Hill
30 PUBLICATIONS   192 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Steven Philip Grover on 25 April 2018.

The user has requested enhancement of the downloaded file.


Brief Review

Tissue Factor
An Essential Mediator of Hemostasis and Trigger of Thrombosis
Steven P. Grover, Nigel Mackman

Abstract—Tissue factor (TF) is the high-affinity receptor and cofactor for factor (F)VII/VIIa. The TF-FVIIa complex is the
primary initiator of blood coagulation and plays an essential role in hemostasis. TF is expressed on perivascular cells and
epithelial cells at organ and body surfaces where it forms a hemostatic barrier. TF also provides additional hemostatic protection
to vital organs, such as the brain, lung, and heart. Under pathological conditions, TF can trigger both arterial and venous
thrombosis. For instance, atherosclerotic plaques contain high levels of TF on macrophage foam cells and microvesicles
that drives thrombus formation after plaque rupture. In sepsis, inducible TF expression on monocytes leads to disseminated
intravascular coagulation. In cancer patients, tumors release TF-positive microvesicles into the circulation that may contribute
to venous thrombosis. TF also has nonhemostatic roles. For instance, TF-dependent activation of the coagulation cascade
generates coagulation proteases, such as FVIIa, FXa, and thrombin, which induce signaling in a variety of cells by cleavage of
protease-activated receptors. This review will focus on the roles of TF in protective hemostasis and pathological thrombosis.
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Visual Overview—An online visual overview is available for this article.   (Arterioscler Thromb Vasc Biol. 2018;38:
709-725. DOI: 10.1161/ATVBAHA.117.309846.)
Key Words: blood coagulation ◼ factor VII ◼ hemostasis ◼ thromboplastin ◼ thrombosis

T issue factor (TF) is a transmembrane protein that func-


tions as a high-affinity receptor for factor (F)VII and
FVIIa.1–6 The TF-FVIIa complex is the primary activator of
TF gene was cloned in 1989, which allowed analysis of its
transcriptional regulation.17 The crystal structure of the extra-
cellular region was found to consist of 2 immunoglobulin-like
the coagulation protease cascade. TF is required to stabi- domains,18 and was the first structure of a member of the class
lize the catalytic site of FVIIa on a plasma membrane (PM) 2 cytokine receptor family. In 1996, 3 groups reported that
to allow optimal interaction with its substrates FIX and FX. deletion of the TF gene in mice leads to universal death during
There are many excellent reviews on TF.1–6 The goal of this embryonic development or shortly after birth, indicating that
review is to summarize our current knowledge on the roles of TF is essential for survival.19–21
TF in hemostasis and thrombosis.
Coagulation Protease Cascade
Brief History of the Major Advances Hoffman and Monroe proposed a new cell-based model of
in TF Research coagulation with a TF-positive cell and activated platelets
The original extrinsic coagulation system consisted of 4 fac- that involves initiation, amplification, and propagation.22 We
tors: fibrinogen (FI), prothrombin (FII), thromboplastin (FIII, have adapted this model and have divided the phases of blood
TF), and Ca++ (FIV). This basic blood coagulation system is coagulation into TF-FVIIa-dependent initiation, inhibition of
present in jawless fish that diverged over 450 million years the TF-FVIIa complex by TF pathway inhibitor (TFPI) and
ago.7 Morawitz was the first to show that conversion of pro- amplification of thrombin generation.
thrombin to thrombin in plasma was induced by tissue throm- After vessel injury or tissue trauma the TF-FVIIa complex
boplastin.8 Indeed, TF was originally simply a laboratory is the spark that triggers blood coagulation by activating both
reagent derived from tissues, such as the placenta, that was FX and FIX. FVII bound to TF is rapidly activated by a vari-
added to plasma to measure the prothrombin time. Early stud- ety of coagulation proteases, although autoactivation by trace
ies showed that TF was a protein that required phospholipid amounts of FVIIa may be the primary pathway.23 Several stud-
for its activity.9,10 It was subsequently purified by 2 groups ies have shown that the TF-FVIIa complex can activate FIX in
in the 1970s.11,12 In 1987, the TF cDNA was cloned by 4 vitro.24–26 One study analyzed the activation of FIX and FX in
groups,13–16 which led to an explosion of research on TF. The an in vitro system and found that FIXa was the major substrate

Received on: December 13, 2017; final version accepted on: January 25, 2018.
From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at
Chapel Hill.
Correspondence to Nigel Mackman, PhD, Division of Hematology and Oncology, Department of Medicine, 111 Mason Farm Rd, 2312C Medical
Biomolecular Research Bldg, CB No. 7126 University of North Carolina at Chapel Hill, Chapel Hill, NC 27599. E-mail nmackman@med.unc.edu
© 2018 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.117.309846

709
710   Arterioscler Thromb Vasc Biol   April 2018

one study reported TF staining throughout thrombotic clots


Nonstandard Abbreviations and Acronyms
but only at the edges of hemostatic clots in mice, and sug-
aPLA antiphospholipid antibody gested that blood-borne TF may contribute to thrombosis but
APS antiphospholipid syndrome not hemostasis.35
EC endothelial cell
F factor TF Expression by Circulating Cells
FVIIai active site–inhibited factor VIIa and Vascular Endothelial Cells
hTF human tissue factor It has been reported that ≈1.5% of CD14+ monocytes in
IVC infrarenal vena cava humans express low levels of intracellular TF under resting
LPS lipopolysaccharide conditions.36 Exposure of monocytes to bacterial lipopolysac-
NAPc2 nematode anticoagulant protein 2 charide (LPS) in vitro or in vivo leads to a rapid and transient
PAR protease-activated receptor expression of TF37–39 (Table 1). This induction is mediated by
PCA procoagulant activity the transcription factors AP-1 and NF-κB.59–61 We think that
PM plasma membrane TF is expressed by monocytes as part of the innate immune
SCD sickle cell disease response presumably to facilitate clot formation to limit the
TF tissue factor dissemination of pathogens.
TFPI tissue factor pathway inhibitor Osterud and Bjorklid62 proposed that TF is exclusively
expressed by circulating monocytes in blood of healthy
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

individuals and that under pathological conditions mono-


of the TF-FVIIa complex.27 Moreover, TF-FVIIa activation of cytes release TF-positive microvesicles that bind to other
FIX is important in vivo because a point mutation at residue cells, such as activated platelets, neutrophils, and endothe-
182, which led to reduced interaction with the TF-FVIIa com- lial cells (ECs). Indeed, in vitro studies support the notion
plex without affecting activation by FXIa, was associated with of the transfer of monocyte-derived microvesicles to plate-
a mild form of hemophilia B.28 In addition to the TF-FVIIa lets and neutrophils.63–66 However, it is difficult to make any
complex, a recent study showed that the TF-FVIIa-FXa com- conclusions about the origins of TF on cells in vivo because
plex can activate FVIII, which would provide additional lev- of the fact that microvesicles can transfer antigens, such as
els of FVIIIa during the initiation phase.29 Formation of the TF, between cells. The most definitive way to determine
prothrombinase complex (FVa-FXa) generates small amounts which cells are making TF is to measure TF mRNA by in
of thrombin in the initiation phase. Activated platelets are an situ hybridization.
important source of FVa that may contribute to the initiation TF expression by platelets, neutrophils, and eosinophils is
phase of blood coagulation.30 The final part of the initiation controversial (Table 1). We found that unstimulated platelets
phase is thrombin activation of the cofactors FV and FVIII contain TF pre-mRNA that is spliced to TF mRNA after acti-
and platelets via protease-activated receptors (PARs). The vation of the platelets, although this process is relatively slow
TF-FVIIa complex is then rapidly inhibited by TFPI in a FXa- (≈30 minutes).42 Other studies have reported variable levels
dependent manner. of TF mRNA and protein in unstimulated platelets but an
In the amplification phase the tenase complex (FVIIIa- increase of TF mRNA and protein in activated platelets.43–45,67
FIXa) is assembled on the surface of activated platelets and Other groups failed to detect TF expression in platelets.49–51
generates additional FXa. The prothrombinase complex is A recent study convincingly showed that a megakaryocyte
also assembled on the surface of activated platelets. Thrombin line expressed TF pre-mRNA, TF mRNA and TF protein and
activation of FXI also allows additional activation of FIX.31 released a subset of TF-positive platelets.46 Interestingly, the
The net result is the generation of large amounts of throm- megakaryocyte cell line contained higher levels of TF mRNA
bin. Thrombin cleaves fibrinogen to fibrin monomers that are versus TF pre-mRNA whereas platelets derived from the
cross-linked by the transglutaminase FXIIIa, which is acti- cells had more TF pre-mRNA. Similar results were observed
vated by thrombin. In addition, thrombin activates additional with megakaryocytes derived from CD34+ cells. It should be
platelets. noted that the megakaryocyte cell line expresses very low
levels of TF (90±15 pg/mL), which is <0.1% of the level of
Blood-Borne TF TF observed in a human pancreatic cell line.68 Several studies
Blood was not thought to contain functional TF because of its have found TF-positive platelets in patients with acute coro-
high procoagulant activity (PCA). However, in 1999, Giesen nary syndromes and diabetes mellitus.47,48 Because of the time
et al32 reported the presence of blood-borne TF that contributed needed to splice TF pre-mRNA and express TF is seems more
to thrombus formation when native blood was flowed over likely that platelet TF contributes to thrombosis rather than
collagen-coated slides. In contrast, Butenas et al33,34 could not hemostasis.69
detect active TF in whole blood. One difference between these TF has been reported to be expressed by neutro-
studies is flow, which may be required to reveal the functional phils, eosinophils, and T-cells in vitro and in vivo
activity of encrypted or intracellular TF. To date, the accumu- (Table 1).52,53,55,56,58 However, the Osterud group failed
lated evidence suggests that there are very low levels of TF in to detect TF expression in human neutrophils or eosino-
blood but this TF is either encrypted or inhibited and must be phils.54,57 We could not detect any functional TF in acti-
activated in some way to activate coagulation. Interestingly, vated human neutrophils (Alexander W and Mackman N,
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   711

Table 1.  Cell Type-Specific Tissue Factor Expression In Vitro increase in TF PCA would enhance the activation of coagula-
and In Vivo tion at sites of vessel injury and cell damage.
Cell Type In Vitro In Vivo
Several mechanisms have been proposed for the post-
translational control of TF PCA.6 The most popular mecha-
Monocyte Yes 37 39,40 nism is that phosphatidylserine interacts with TF resulting in a
Endothelium Yes 41 40 conformational change and enhanced PCA on cell surfaces.88
Platelets Yes 42–46 47,48 In support of this mechanism, TF PCA is enhanced by the
addition of phosphatidylserine to reconstituted phospholipid
No 49–51 …
vesicles.86,89 Additionally, the pseudosubstrate TFPI-FXa
Neutrophils Yes 52 53 bound to the TF-FVIIa complex on perturbed cells exposing
No 54 … phosphatidylserine but not to the complex on unperturbed
cells, which suggested that activation of the cells leads to a
Eosinophils Yes 55,56 …
change in conformation of TF that enables engagement with
No 57 … substrate.88 Figure 1A shows a summary of the proposed phos-
T-cells Yes 58 58 phatidylserine-dependent model of TF de-encryption. Briefly,
the PM of eukaryotic cells has an asymmetrical distribution
of phospholipids with negatively-charged phospholipids, such
as phosphatidylserine and phosphatidylethanolamine, seques-
unpublished data, 2013). However, there is stronger evi- tered in the inner leaflet and neutrally charged phospholipids,
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

dence for TF expression by neutrophils in monkeys, rabbits, such as phosphatidylcholine and sphingomyelin, present in the
and mice compared with humans70–73 (Erlich J and Mackman outer leaflet. Cellular damage leads to loss of the PM asym-
N, unpublished data, 2000). In addition, we found that dele- metry and exposure of negatively-charged phospholipid, such
tion of TF in neutrophils reduces LPS-induced activation as phosphatidylserine, on the cell surface. In a recent study,
of coagulation in mice (Grover and Mackman, unpublished molecular dynamic simulations were used to generate a model
data, 2018). of membrane-bound TF-FVIIa complex using soluble TF,
TF expression by ECs is readily demonstrably in vitro but which lacks the transmembrane domain.90 It was found that
it is more difficult to demonstrate TF expression by ECs in “the direct interaction of the phosphatidylserine headgroups
vivo in part because of the very low levels of expression.74–76 with specific residues on soluble TF most likely also affect the
Typically, studies are performed with cultured ECs, such as conformation of these residues.” Further, they concluded that
human umbilical vein ECs. It is often assumed that these cells “lipids might have an active direct role in de(-en)cryption of
can be used as models of the variety of ECs present in differ- soluble TF.”90 Phosphatidylserine exposure on the cell surface
ent vascular beds in vivo, which is unlikely to be true. We and also facilitates the binding of coagulation proteins (FII, FVII,
others have shown that human umbilical vein ECs express TF FIX, and FX) with positively-charged γ-carboxyglutamic acid
in response to a variety of different agonists and this induction (Gla) domains and assembly of the cofactor/protease com-
is mediated by activation of NF-κB.41,77,78 The most convincing plexes. In contrast to phosphatidylserine-rich lipid surfaces,
examples of TF expression in vivo are by ECs in the splenic phosphatidylcholine-rich lipid surfaces confer relatively lim-
microvascular, at arterial branching areas in septic baboons ited TF PCA suggesting that phosphatidylcholine maintains
and in the lungs of sickle cell disease (SCD) mice.79–82 Another TF in an encrypted state.91,92
study reported TF expression in ECs in the glomeruli of endo- More recently it was reported that sphingomyelin, a major
toxemic mice.83 One difficulty with these immunohistochemi- phospholipid in the outer leaflet of the PM, helps maintain
cal studies is TF staining of a given cell does not mean that the the TF-FVIIa complex in an encrypted state in resting cells.93
cell expressed TF because this signal could be because of the Cellular activation leads to translocation of acid sphingomy-
binding of TF-positive microvesicles from other cells, such as elinase to the PM and hydrolysis of sphingomyelin enabling
activated monocytes. de-encryption of the TF-FVIIa complex (Figure 1A).
TF PCA is also regulated by oxidation of an alloste-
Transcriptional and Post-Transcriptional ric disulfide bond (Figure 1B). TF has 2 pairs of disulfide
Regulation of TF bonds, one in the N-terminal (Cys49-Cys57) and the other
Because of the large amplification of the coagulation cas- in the C-terminal (Cys186-Cys209) domain. An early study
cade, TF-dependent activation of blood coagulation needs demonstrated that TF PCA was reduced ≈3-fold when
to be tightly regulated to maintain hemostasis and prevent either Cys186 or Cys209 were mutated whereas there
thrombosis. The first level of control is at the level of TF gene was no effect of mutating either Cys49 or Cys57.94 This
expression. For instance, high TF expression is restricted to suggested that formation of the Cys186-Cys209 disulfide
cells not in direct contact with blood.79,84 Additionally, TF bond was important for TF PCA. A later study proposed
PCA is regulated at the post-translational level. TF exists in that cryptic TF contains unpaired cysteine thiols at posi-
2 states: encrypted TF present on unperturbed cells and de- tions 186 and 209, and that activation involves formation
encrypted TF on damaged cells.85 FVIIa can bind to TF on of a Cys186-Cys209 disulfide bond.95 Protein disulfide
cells under resting conditions but the level of TF PCA activity isomerase has been proposed to be an important regula-
is very low. However, activation or disruption of cells dramati- tor of TF redox state and activity.96–98 For instance, inhi-
cally increases TF PCA.86,87 In terms of hemostasis, this rapid bition of protein disulfide isomerase reduces TF PCA in
712   Arterioscler Thromb Vasc Biol   April 2018
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Figure 1. Mechanisms of tissue factor (TF) de-encryption. A, The distribution of phospholipids in the plasma membrane of resting cells
is asymmetrical and contains phosphatidylcholine (PC) and sphingomyelin (SM) primarily in the outer leaflet and phosphatidylinositol (PI),
phosphatidylserine (PS) and phosphatidylethanolamine (PE) in the inner leaflet. The presence of SM and PC in the outer leaflet maintains
TF in an encrypted state. Upon cell activation phospholipid scramblase mediates transfer of PS to the outer leaflet. Cellular activation also
leads to release of acid sphingomyelinase (ASM) that catalyzes the conversion of SM to ceramide (Cer). These 2 processes lead to de-
encryption of TF increasing its cofactor activity and enabling efficient activation of FX by the TF-FVIIa complex. B, Encrypted TF contains
2 free thiols at Cys186 and Cys209. It is proposed that protein disulfide isomerase (PDI) catalyzes the formation of an allosteric disulfide
bond (Cys186-Cys209) that de-encrypts TF and enhances its procoagulant activity.

vitro.97,99 However, there are several unresolved issues with bind to the TF-FVIIa complex, recombinant TFPI, active site-
the allosteric disulfide bond hypothesis. First, the disul- inhibited FVIIa (FVIIai), nematode anticoagulant protein C2
fide bond must be formed before binding of FVII/FVIIa (NAPc2), and Ixolaris. NAPc2 is an 85-amino acid protein
to TF because this region of TF is buried at the interface isolated from the hematophagous hookworm.103 It acts in a
between TF and FVII/FVIIa.100 Second, as noted by Bach similar manner to TFPI by first binding to FXa and then the
and Monroe100 “there is no direct quantitative evidence for NAPc2-FXa complex binds to TF-FVIIa. Ixolaris was isolated
free thiols in TF.” Finally, protein disulfide isomerase may from a tick salivary gland and contains 2 Kunitz domains.104
also indirectly affect TF PCA by promoting exposure of Unlike TFPI, it binds to zymogen FX and then the complex
phosphatidylserine.101 It should be noted that studies of de- inhibits the TF-FVIIa complex.
encryption of TF PCA have largely used cultured cells and These inhibitors have been used in preclinical and
it is difficult to know what mechanism(s) occur in vivo. clinical studies of acute coronary syndromes, sepsis, HIV
infection, and acute lung injury/acute respiratory distress
TF Inhibitors syndrome. The effect of NAPc2 was examined in patients
After the cloning of the TF cDNA in the late 1980s there was with non–ST-segment–elevation acute coronary syndromes
a lot of excitement about the prospect of using TF inhibitors and seems to be safe because it did not significantly affect
as new antithrombotics.102 This is summarized by a com- major or minor bleeding (3.7% versus 2.5%).105 However, no
ment by Dr Golino in 2002 “many experimental studies have further studies have been performed using NAPc2. A recent
demonstrated that inhibitors of TF:FVIIa PCA are powerful study showed that Ixolaris reduced levels of plasma D-dimer
inhibitors of in vivo thrombosis and that this approach usually in monkeys infected with simian immunodeficiency virus.106
results in less pronounced bleeding tendency, as compared Recombinant TFPI reduced lethality in a baboon model of
with other more classical antithrombotic interventions.”102 sepsis.107 However, there was no effect on all-cause mortal-
There are 5 main classes of human TF (hTF) inhibitors: anti- ity in patients with severe sepsis and there was an increased
TF antibodies that either compete with FVII/VIIa binding or risk of bleeding.108 Early administration of a monoclonal
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   713

antibody against TF reduced sepsis-induced respiratory


and renal failure in baboons.109 The success of this preclini-
cal model led to a clinical trial that examined the effect of
FVIIai in patients with acute lung injury/acute respiratory
distress syndrome. Unfortunately, the trial was stopped
early by the Safety Committee because of increased mortal-
ity in the group receiving the highest dose of FVIIai.110,111 In
addition, patients with the higher doses of FVIIai also had a
trend to increased risk of serious bleeding. In retrospect, given
the central role of TF in lung hemostasis, these results are not
unexpected. Based on these studies, it seems unlikely that TF
inhibitors will be used clinically in the future.

TF-FVIIa-Dependent Signaling
The TF-FVIIa complex has both PCA and signaling activity.
The localization of TF on the cell surface seems to regulate
these functions with the signaling form of TF being located
in lipid rafts.112 The signaling form of the TF-FVIIa complex
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

also interacts with β1 integrins.113 A recent study identified the


binding motif in the FVIIa protease domain that mediates the
interaction with β1.114
The TF-FVIIa complex and downstream coagulation
proteases, such as thrombin, activate cells by cleavage of G Figure 2. Role of different coagulation proteins in hemostasis
protein-coupled PARs. This family consists of 4 members, and thrombosis. Based on the hemostatic defects of mice with
complete deficiencies in the different coagulation factors, we can
PAR1–4.115 PAR2 can be activated by the TF-FVIIa and divide the coagulation factors into 4 groups. Group 1 consists of
TF-FVIIa-FXa complexes as well as FXa, whereas PAR1 is factors in the extrinsic pathway (tissue factor [TF], factor [F] VII,
activated by thrombin and FXa.116 TF-FVIIa-dependent sig- FV, FX, FII). Group 2 consists of FIX and FVIII. Group 3 consists
of FXI. Group 4 consists of FXII. Pathological TF expression and
naling has been mainly studied in cancer.117 Studies have FXII can trigger thrombosis.
shown that PAR2 activation by TF-FVIIa induces proangio-
genic and immune modulating cytokines, whereas PAR1 acti- model.131–135 Group 3 contains FXI. A complete deficiency of
vation by thrombin or the TF-FVIIa-FXa complex contributes this protein in mice is not lethal but these mice have a mild
to the metastatic tumor environment.117 hemostatic defect in a saphenous vein injury model.135,136
Group 4 contains FXII. In contrast to the hemostatic defects
Alternatively Spliced TF in mice with deficiencies in proteins in the other groups, mice
A soluble form of TF can be generated by alternative splicing with a complete deficiency in FXII have no hemostatic defects
of the TF gene where exon 5 is skipped.118,119 This alternatively (Figure 2).137 However, FXII-deficient mice are protected in
spliced TF lacks residues required for interaction with FX, the thrombosis models.138,139
allosteric disulfide in the C-module and the transmembrane Unlike mouse studies where gene inactivation leads to a
domain that dramatically reduce its PCA.120–122 However, complete deficiency of a given protein, humans with deficien-
alternatively spliced TF has PCA in the presence of phospho- cies in different coagulation proteins usually have residual
lipids.123 Moreover, alternatively spliced TF plays a role in amounts of functional protein. To date, there are no reports
cancer biology by promoting angiogenesis and proliferation of humans with a deficiency of TF. In contrast, rare inherited
of tumor cells via interacting with integrins containing β1 and bleeding disorders occur because of deficiencies in FII, FVII,
β3.124,125 FX, FXI, and FXIII,140 although the estimated prevalence of
these disorders is very low (between 1 and 2 per 106 individu-
als).140 Individuals with deficiencies in FII, FX, or FV most
Hemostasis
often bleed in the skin and mucus membranes and those with
Hemostatic Defects in Mice and Humans the lowest levels of the different coagulation factors have intra-
Deficient in Different Coagulation Proteins cranial bleeds.140 In terms of the coagulation cascade, the most
Factors of the coagulation cascade can be divided into 4 groups frequent congenital bleeding disorders in the general human
based on the phenotype of mice deficient in each of the factors population are caused by deficiencies in FVIII (hemophilia A)
(Figure 2). Group 1 contains factors of the extrinsic pathway and FIX (hemophilia B). The prevalence rates of these bleed-
(TF, FVII, FV, FX, and FII). Complete deficiencies of any of ing disorders is 1/5000 male births.140 Hemophilia A and B
these proteins in mice leads to universal death during embry- patients are prone to spontaneous bleeding in the joints and
onic development or shortly after birth.19–21,126–130 Group 2 con- skeletal muscle.141 FXI deficiency is rare (1 per 100 000) and
tains FVIII and FIX. Complete deficiencies of these proteins individuals with FXI deficiency rarely exhibit spontaneous
in mice is not lethal but is associated with impaired hemo- bleeding but have an elevated risk of bleeding after surgery
stasis in a tail transection model and a saphenous vein injury such as tooth extraction.142–144
714   Arterioscler Thromb Vasc Biol   April 2018

TF as a Hemostatic Envelope Around Blood Table 2.  Hemostatic Defects in Low Tissue Factor Mice
Vessels and Body Surfaces Phenotype References
The TF-FVII complex plays an essential role in hemostasis.
Spontaneous
TF is expressed by perivascular cells, such as adventitial fibro-
blasts and pericytes, and at body surfaces, such as keratino-  Lung Fatal hemorrhage and 154,155
cytes in the skin.79,84,145 Drake et al79 proposed that TF formed a hemosiderin
hemostatic envelope around blood vessels and body surfaces.  Brain Fatal hemorrhage 152,154
Initially, it was thought that the physical separation of TF from  Uterus Fatal postpartum hemorrhage 156
blood meant that the TF-FVIIa complex was only formed after
 Placenta Blood pools in the placenta 156
vascular injury. However, a recent study reported that perivas-
cular TF was prebound to FVII/FVIIa in the dermal vascu-  Testis Hemorrhage and calcification 2,154
lature.146 Indeed, coagulation factors, such as FVII, leak into  Heart Hemorrhage and fibrosis 157,158
the extravascular space at levels roughly proportional to their
 Intestine Hemorrhage 152,154
size.147,148 The presence of a preformed TF-FVIIa complex
would allow rapid activation of clotting after vessel injury. In Induced
contrast to dermal vessels, there is a high proportion of free TF  Intratracheal Increased hemorrhage 159
(non-FVII bound) present in the brain parenchyma and cere- lipopolysaccharide
bral vessels.149 This difference is likely because of the lower   Influenza A infection Increased hemorrhage 160
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

permeability of the blood–brain barrier. The high levels of TF


 Collagenase-induced Increased hemorrhage 73
in the brain may compensate for this the lower level of pre-
intracerebral hemorrhage
formed TF-FVIIa complex around the cerebral vasculature.
  Tail vein transection Prolonged bleeding 152

Use of Transgenic Mice to Study  Saphenous vein laser Prolonged bleeding 161
injury model
the Role of TF in Hemostasis
As noted above, TF−/− and FVII−/− mice die during embryonic  Saphenous vein Prolonged bleeding Unpublished
development or shortly after birth, which means that they mechanical injury model
cannot be used to study the roles of the TF-FVIIa complex   Skin punch Delayed wound healing 162
in hemostasis in different organs in adult mice.19–21,126 To cir-
cumvent the lethal phenotype of these mice, we and others
have generated mice that express low levels of either TF or FXI−/− mice survive to wean (Grover and Mackman, unpub-
FVII.150,151 We rescued mTF−/− embryos by expressing hTF lished data, 2018).
from a hTF minigene.150 These mice (mTF−/−, hTF+ minigene) Mice with low levels of TF but lacking PAR4 survive to
are referred to as low TF mice and express ≈1% to 2% of wild- wean but exhibit reduced survival because of fatal bleeds in
type TF levels. Rosen et al151 generated transgenic mice with the lung and brain compared with low TF, PAR4+/+ controls.154
low levels of FVII by targeted replacement of the FVII gene Interestingly, the bleeding phenotype observed in low TF mice
with a FVII cDNA. Remarkably, the phenotypes of low TF in the laser injury model was increased after administration
and low FVII mice are similar supporting the notion for a cen- of the P2Y12 inhibitor clopidogrel at a dose that had a mini-
tral role for the TF-FVIIa complex in hemostasis.152,153 mal effect on bleeding time in wild-type mice.161 These data
Consistent with a role for TF in hemostasis, low TF mice suggest that TF-dependent thrombin generation leads to both
exhibit prolonged bleeding after various hemostatic chal- fibrin deposition and platelet activation that act co-operatively
lenges. For instance, low TF mice have increased bleed- to form and stabilize the hemostatic plug.
ing after tail transection (Table 2).152 In addition, low TF Two other transgenic mouse lines have been made that
mice had prolonged bleeding times in the saphenous vein express hTF in place of mouse TF. Chromosomal vector hTF
mechanical injury model and after repeated vessel injury in mice (mTF−/−, hTF+ chromosomal vector) express hTF from
the saphenous vein laser injury model161 (Table 2; unpub- a vector consisting of the majority of chromosome 1, whereas
lished data, 2018). Interestingly, the hemorrhage observed in hTF knockin mice were generated by gene targeting that
low TF mice in these models is not as severe as FIX−/− mice. replaced the mouse TF gene with the hTF gene.157,164 Mouse
Low TF mice also have excess bleeding after a full thickness FVIIa binds with high affinity to both mouse and hTF but the
skin punch.162 association of human FVIIa with mouse TF is significantly
We determined the effect of combining low levels of TF lower than with hTF.165 Indeed, we found a 6-fold reduction
with a complete deficiency of other procoagulant factors or in FXa generation when we used human FVIIa with mouse TF
PAR4, which is the main thrombin receptor on mouse plate- compared with hTF.166 These studies indicate that hTF can be
lets. Consistent with the importance of FIX in hemostasis, used to replace mouse TF but not vice versa, although we do
mice with a combination of low levels of TF and an absence not know about interactions of hTF with mouse β1 integrins.
of FIX do not survive to wean.163 Our recent studies indicate Finally, we generated mice with a floxed TF gene (TFfl/fl) that
that low TF, FIX−/− mice survive embryonic development but when crossed with mice expressing Cre recombinase under the
die shortly after birth (Grover and Mackman, unpublished control of a specific promoter leads to cell type-specific dele-
data, 2018). In contrast, we found that the majority of low TF, tion of the TF gene (Table 3).157
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   715

Table 3.  Transgenic Mice With Tissue Factor Deleted in Tissue-Specific Hemostasis
Different Cell Types TF is expressed in an organ-specific manner with high levels
Cre Line Cell Type Phenotype References in the brain, lung, heart, uterus, and placenta, and low levels
in skeletal muscle and joints.183 This pattern of expression
Mlc2v Cardiomyocyte Cardiac fibrosis 157
suggests that TF provides additional hemostatic protection
SM22 Vascular smooth Reduced carotid arterial 167 to a select group of vital organs. For instance, blood ves-
muscle cell thrombosis
sels in the heart and lung may need additional hemostatic
LysM Myeloid cell Reduced 168 protection because they are more likely to be injured by the
lipopolysaccharide- mechanical movement of these organs. In contrast, tissues
induced coagulation that have low levels of TF are likely more dependent on
Tie2 Endothelial and Reduced 168 the intrinsic pathway to generate thrombin for hemostasis.
hematopoietic cell lipopolysaccharide- Indeed, hemophilia A and B patients are prone to bleeding
induced coagulation into joints and skeletal muscle and inhibitor patients receive
Alb Hepatocyte Reduced acute 169,170 FVIIa as a bypass agent to boost the extrinsic pathway. Low
liver injury induced TF mice have been used to study the role of TF in tissue-
coagulation specific hemostasis and exhibit spontaneous hemorrhaging
SPC Lung epithelial Reduced acute 160,171 in various tissues (Table 1).
lung injury induced
coagulation
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Lung
Nestin Neuroectodermal Increased collagenase- 172 The lung contains high levels of TF.184 Epithelial cells are
induced intracerebral the primary site of TF expression in the lung.61 Mice with TF
hemorrhage
deleted in epithelial cells (TFfl/fl, SPCcre; Table 3) have sig-
K14 Keratinocyte Delayed wound healing In nificantly reduced baseline levels of TF compared with con-
preparation trols.160,171 LPS and influenza A virus infection increase TF
expression in the lung of mice and specifically in epithelial
Hemostasis Requires a Balance cells.61,159,160,171,184 LPS and influenza A virus did not increase
Between TF and TFPI TF expression in TFfl/fl, SPCcre mice. These studies indicate
TFPI is the major physiological inhibitor of the TF-FVIIa com- that the increased TF expression in epithelial cells is response
plex and shuts down the initiation of the clotting cascade.173–177 to infection may provide additional hemostatic protection in
There are 2 forms of TFPI: TFPIα and TFPIβ. TFPIα contains the face of an infection.
3 Kunitz domains where domain 1 binds to FVIIa and domain Low TF mice exhibit a high incidence of fatal lung hem-
2 binds to FXa. TFPIβ is alternatively spliced form that con- orrhages (≈50%) when backcrossed 7 generations onto the
tains Kunitz 1 and 2 with a different C-terminus that mediates prohemorrhagic C57BL/6 background. (Table 2).154,155 This
anchoring to the membrane via a glycosylphosphatidyl inosi- indicates that lung hemostasis is impaired in low TF mice.
tol linkage. Similar to a TF−/− mice, a complete deficiency of In addition, our standard low TF (backcrossed 6 genera-
TFPI is universally lethal during embryonic development.178 tions to the C57BL/6 background) exhibit increased alveolar
TFPI−/− embryos had intravascular thrombi and fibrin(ogen) hemorrhage compared with control mice when challenged
deposition in the liver and seem to exhibit a consumptive with intratracheal LPS (Table 2).159 As expected, both low
coagulopathy with secondary bleeding. There are no reports TF and TFfl/fl, SPCcre mice had increased alveolar hemor-
of TFPI-deficient humans. rhage after influenza A virus infection (Tables 2 and 3).160
The effect of reducing the level of FVII on the survival However, significantly more bleeding was observed in low
of TFPI−/− embryos was evaluated by intercrossing FVII+/−, TF mice compared with TFfl/fl, SPCcre mice, and we hypoth-
TFPI+/− mice.179 FVII+/−, TFPI−/− mice were rescued from esize that additional cell types, such as fibroblasts, also con-
embryonic lethality but died shortly after birth because of tribute to hemostasis in the lung. We are currently testing this
consumptive coagulopathy. hypothesis.
Similarly, FVII−/−, TFPI−/− survived embryonic develop-
ment but succumbed to perinatal bleeding because of an Brain
absence of FVII. We found that low levels of TF rescued The brain contains high levels of TF with astrocytes being
TFPI−/− embryos from embryonic lethality and allowed the the major source.184,185 Low TF mice occasionally succumb
generation of viable TFPI−/− adult mice that survived for to spontaneous fatal brain hemorrhages (Table 2).152,154 In
>6 months without any adverse phenotype.155 Conversely, addition, administration of an antihuman TF antibody to TF
reducing the levels of TFPI dose-dependently reduced the knockin mice expressing hTF resulted in cerebral hemor-
hemostatic defects in low TF mice.155 A similar principle of rhage.164 Importantly, FVII−/− and FII−/− neonates also have
rebalancing clotting has been applied to restore hemostasis in fatal cerebral hemorrhages.126,129,130 In addition, genetic elimi-
hemophilic mice. For instance, inhibition of TFPI in hemo- nation of FII in adult mice led to spontaneous hemorrhage in
philia A mice reduced bleeding in a tail vein model.180 TFPI the brain.186,187
inhibitors are currently being developed for the treatment of Recently, the Pawlinski group generated mice with TF
patients with hemophilia.181,182 deleted in all neuronal cells (TFfl/fl, nestin-cre; Table 3).172
716   Arterioscler Thromb Vasc Biol   April 2018

These mice express a very low level of TF (≈1%) in their Table 4.  Cellular Sources of Tissue Factor That Drive
brains compared with a slightly higher level of TF (≈2%) in Thrombosis and Disseminated Intravascular Coagulation
the brains of low TF mice. A model of collagenase-induced Cellular Source
intracerebral hemorrhage was used to determine the role of Disease Model of Tissue Factor References
TF in brain hemostasis. Both low TF and TFfl/fl, nestin-cre
Atherosclerosis Macrophage foam cells and 191–193
mice exhibited increased intracerebral hemorrhage com- macrophage-derived microvesicles
pared with controls.172 However, more hemorrhage was
Sepsis/endotoxemia Monocytes 39,40,168
observed in TFfl/fl, nestin-cre compared with low TF mice
and this was associated with death of some of these mice. Surgery Peripheral blood mononuclear cells 194
These studies suggest that the high levels of TF in the brain Cancer Tumor cell-derived microvesicles 195–197
are required to limit intracerebral hemorrhage after trau- Antiphospholipid Monocytes 198–200
matic brain injury. syndrome
Liver injury Hepatocytes 169,170
Heart Sickle cell disease Monocytes 73,201
TF is expressed by both cardiac fibroblasts and cardiac myo-
Viral infection Peripheral blood mononuclear 202,203
cytes in the heart.184 Administration of an inhibitory anti-
cells/monocytes
human TF antibody to TF knockin mice expressing HTF
resulted in cardiac hemorrhage.164 We have observed occa-
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

sional spontaneous hemorrhages in the heart of low TF mice


and hearts of older low TF mice are fibrotic.158 Mice with Atherosclerosis
only ≈20% expression of hTF in the heart and those with In humans, rupture of advanced atherosclerotic lesions trig-
TF deleted in cardiac myocytes (TFfl/fl, Mlc2v; Table 3) also gers arterial thrombosis leading to myocardial infarction
exhibit cardiac fibrosis.157 Importantly, a similar pattern of and stroke. Atheroma contain high levels of TF that likely
cardiac fibrosis has been observed in low FVII, low FX, and plays a central role in atherothrombosis (Figure 3A).191,204–210
low FII expressing mice but not FIX deficient mice.133,153,188,189 Macrophage foam cells are the main source of TF in plaques
Spontaneous hemorrhage was also observed in the heart with apoptotic macrophages releasing TF-positive microves-
after genetic elimination of prothrombin in adult mice.186 icles into the necrotic core.192,193,211 In addition, patients with
These results indicate that the extrinsic pathway, but not the acute coronary syndromes have increased levels of TF expres-
intrinsic pathway, mediates cardiac hemostasis.153,158,186,188,189 sion in monocytes, platelets, and leukocyte-platelet aggre-
gates that may also contribute to thrombosis after plaque
rupture.48,212 Many studies have reported increased levels of
Uterus, Placenta, and Testis
TF antigen in the plasma of cardiovascular disease patients.213
Female low TF mice exhibit excess bleeding after delivery and
However, we could not detect increased levels of microvesicle
14% of the mice die, which indicates that TF is required for
TF activity in these patients.214
uterine epithelium hemostasis postpartum (Table 2).156Low
In mice a 50% reduction of TFPI increased thrombo-
TF placentas contain blood pools in the labyrinth (Table 2).156
sis after photochemical disruption of carotid artery plaques
At present, it is unclear whether this is because of a hemo-
in ApoE−/− mice, presumably because of an increase in TF
static defect or a structural defect in the placental labyrinth.
PCA.215 Importantly, inhibition of TF with FVIIai markedly
Interestingly, FVIII−/− and FIX−/− mice have no hemostatic reduced thrombosis after ultrasonic disruption of carotid
defects in the uterus or placenta.132,133 plaques in ApoE−/− mice.216 Similarly, FVIIai or recombinant
We observed that male low TF mice lost fecundity at a TFPI reduced thrombosis after balloon-mediated disrup-
relatively young age and had shrunken and fibrotic testes. tion of carotid plaques in hypercholesterolemic rabbits.217,218
We also observed spontaneous hemorrhage in the testes of These studies indicate that TF within atherosclerotic plaques
low TF mice that may have been induced by trauma or dur- plays a major role in the formation of occlusive thrombi after
ing retraction of the testes into the abdomen. We think that rupture.219
hemorrhage leads to fibrosis of the testes and the loss of
fertility. Sepsis/Endotoxemia
Disseminated intravascular coagulation is a life-threatening
Thrombosis disorder caused by diffuse activation of coagulation that
Thrombosis is a pathological process that occludes blood flow can result in microvascular occlusion and organ dysfunc-
in vessels. TF is involved in the pathogenesis of both arte- tion.220 Disseminated intravascular coagulation is frequently
rial and venous thrombosis.190 Sources of TF that activate observed in patients with bacterial sepsis. TF has been shown
coagulation in different diseases include macrophages and to play a central role in disseminated intravascular coagulation
microvesicles in atherosclerotic plaques, activated cells within (Figure 3B).221 In a human model of LPS-induced endotoxemia
the vasculature and perivascular cells that are exposed because TF mRNA and TF protein expression were induced in blood
of an increase in vascular permeability. cells (presumably monocytes) and this was associated with
There is strong evidence that monocyte TF contributes to increased levels of thrombin-antithrombin complex, a marker
thrombosis in many diseases (Table 4). of activation of coagulation.39 TF expression is also observed
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   717
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Figure 3. Cellular sources of tissue factor (TF) that trigger thrombosis and disseminated intravascular coagulation. A, TF within athero-
sclerotic plaques triggers thrombosis after plaque rupture. Other sources of TF in patients with cardiovascular disease include circulating
activated monocytes and activated platelets. B, In endotoxemia and sepsis, pathogens induce TF expression in monocytes and endothe-
lial cells and the release of TF-positive microvesicles (MVs). In addition, increased vascular permeability exposes TF on perivascular cells
to blood. All these sources of TF may contribute to disseminated intravascular coagulation. C, Pancreatic tumors express high levels of
TF and release TF-positive MVs that circulate in the blood and trigger venous thrombosis. D, Antiphospholipid antibodies (aPLA) induce
TF expression in monocytes and the release of TF-positive MVs that trigger thrombosis. TF expression by endothelial cells may also con-
tribute to local thrombosis. Administration of the antioxidant ubiquinol reduced Coenyzme Q10 (CoQ10) reduces monocyte TF expression.

in monocytes isolated from septic baboons.40 As noted earlier, of the anticoagulant proteins EC protein C receptor and
immunohistochemistry studies demonstrated TF staining on thrombomodulin.83 Taken together, we propose that activated
ECs in the spleen and aorta of septic baboons and ECs in the monocytes are the major source of TF that activates coagula-
glomerulus of endotoxemic mice.38,80,83 Therefore, EC TF may tion during endotoxemia and sepsis. However, we speculate
also play a role the activation of coagulation during sepsis and that TF expressed on perivascular cells exposed by increased
endotoxemia. Importantly, early inhibition of TF reduced the vascular permeability and possibly TF induced on ECs also
activation of coagulation and mortality in septic baboons.107,222 contribute to the activation of coagulation in sepsis.
We analyzed the role of TF in the activation of coagulation
in a mouse model of endotoxemia. As expected, LPS activa- Surgery
tion of coagulation was lower in low TF mice compared with Surgery is associated with an increased risk of venous throm-
controls.223 Similarly, wild-type mice containing bone marrow bosis. Three studies have reported an increase in monocyte
from low TF mice or mice with TF deleted in myeloid cells TF expression 1 day after surgery to remove tumors as well
(TFfl/fl, LysMCre; Table 2) had lower levels of activation of as after coronary artery bypass surgery and total knee arthro-
coagulation compared with controls.168 However, bone mar- plasty.194,224,225 This suggests that surgery triggers an inflamma-
row cells only account for ≈50% of TF that activates coagula- tory response surgery that induces monocyte TF expression.
tion in endotoxemic mice. We found that deleting TF on ECs Importantly, the increase in monocyte TF occurs before the
did not affect activation of coagulation in a mouse endotox- peak of clinical venous thrombosis, which is 7 days after sur-
emia model.168 Another study found that inducible expression gery.226 These studies suggest that induction of TF expression
of a degradation resistant form of IκBα in ECs blocked the on monocytes contributes to postsurgery venous thrombosis.
induction of TF expression in ECs in the glomerulus of endo-
toxemic mice.83 However, the specific impact of this blockade Cancer
of EC TF expression could not be evaluated because expres- Thromboembolism represents a leading cause of death in
sion of the mutant IκBα also prevented the downregulation patients with cancer and is associated with poor prognosis.227
718   Arterioscler Thromb Vasc Biol   April 2018

Venous thrombosis, in particular, is frequently observed The exact mechanism by which aPLA induce a hyperco-
in cancer patients with reported annual incidences ranging aguable state remains unclear. However, results from differ-
between 1.5% to 20% per annum depending on the site of can- ent studies suggest that aPLA induction of TF expression in
cer and treatment.228 In vitro, cancer cell lines shed TF-positive monocytes may underpin the pathogenesis of APS-associated
microvesicles.68,229–231 Moreover, increased levels of circulat- thrombosis. Importantly, human monocytes isolated from
ing TF-positive microvesicles are observed in patient with dif- patients with APS express TF.198–200 Moreover, incubation
ferent types of cancer, including pancreatic, brain, colorectal, of primary human monocytes from healthy individuals with
lung, breast, and prostate.195,232–235 Importantly, elevated levels anti-β2-glycoprotein I IgG leads to increased monocyte TF
of MV TF activity are associated with venous thromboembo- gene expression, PCA and release of TF-positive microves-
lism in pancreatic cancer patients but not in brain, gastric, or icles.243–246 In vitro studies showed that aPLA induces oxi-
colorectal cancer patients.195–197,228 dative stress in monocytes, and levels of cell surface TF
Preclinical studies of cancer-associated thrombosis have exposure correlate with intracellular peroxide generation.247
largely focused on pancreatic cancer, given the high rates of Furthermore, pretreatment of the monocytes with the antioxi-
thrombosis observed in patients with this type of malignancy. dant ubiquinol, reduced Coenzyme Q10, decreased oxidative
Immunocompetent and immunodeficient mouse models have stress and TF expression.248 In a follow-up study, adminis-
been used to study the role of tumor-derived, TF-positive tration of Coenzyme Q10 to APS patients reduced oxidative
microvesicles in thrombosis (Figure 3C). Studies have stress and TF exposure on monocytes.248,249 Further clinical
used the mouse pancreatic cell line Panc02 that expresses studies are required to determine if antioxidant therapies can
TF. Injection of mice with Panc02-derived, TF-positive effectively prevent thrombotic events in patients with APS by
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

microvesicles enhanced thrombosis in a laser injury model reducing monocyte TF expression.


and an infrarenal vena cava (IVC) stenosis model, whereas Animal models have been used to gain insights into
microvesicles from Panc02 cells with reduced TF expres- the involvement of TF in APS-associated thrombosis
sion were less thrombotic.236,237 Moreover, immunocompetent (Figure 3D).250,251 For instance, low TF mice were protected
mice bearing Panc02 tumors had enhanced thrombus forma- from the thrombotic microangiopathy induced by the pas-
tion in a mesenteric ferric chloride model and an IVC stenosis sive transfer of a murine monoclonal aPLA or purified
model.236,237 patient aPLA.252 Interestingly, aPLA were shown to induce
Immunodeficient mice are required for studies with TF expression in monocytes and ECs via a pathway involv-
human pancreatic cancer cell lines. We and others have ing endosomal NADPH oxidase and TLR7/8.253 Importantly
used several TF-expressing human pancreatic cells lines, induction of TF was absent in aPLA-treated monocytes from
including L3.6pl, HPAF-II, and BxPc-3.68,238,239 Injection NOX2−/− mice. A follow-up study with mice showed that
of TF-positive microvesicles derived from these cell lines aPLA enhanced thrombosis in an IVC stenosis model.254
activated coagulation and increased thrombus formation Enhanced thrombosis in this study required NOX2 and leu-
in the IVC stenosis model.68,238,239 In addition, inhibition of kocytes but not TLR4 or platelets.250 These 2 studies sug-
TF on BxPc-3 microvesicles with an antihuman TF anti- gest a role for monocyte TF in aPLA induced thrombosis.254
body abolished the enhanced thrombosis. Mice bearing Further studies are needed to understand how TF contributes
HPAF-II tumors also demonstrated markedly shortened to thrombosis in patients with APS.
time to occlusion in a saphenous vein ferric chloride injury
model but there was no increase in thrombosis in the IVC Acute Liver Injury and Cirrhosis
stenosis model.68 However, mice bearing BxPc-3 tumors Acute liver injury and cirrhosis are associated with a pro-
had an increase in thrombosis in the IVC stenosis model.238 thrombotic state. One study found very high levels of circulat-
Recently, we evaluated the contribution of tumor derived, ing MV TF activity in patients with acute liver injury.255 We
TF-positive microvesicles to venous thrombus in tumor found that MV TF activity was increased with cirrhosis and
bearing mice.240 Mice with orthotopic BxPc-3 tumors had levels were associated with the Child–Pugh score and markers
larger thrombi in an IVC stasis model compared with con- of activation of coagulation and fibrin degradation.256
trols and administration of an antihuman TF antibody sig- The liver expresses low levels of TF compared with other
nificantly reduced the size of the thrombi.240 Taken together, organs, such as the brain and lung. The Luyendyk group deter-
these studies suggest that tumors release TF-positive mined the contribution of hepatocytes to basal TF expression
microvesicles into the circulation and that these microvesi- in the liver by comparing levels in mice with TF deleted in
cles enhance venous thrombosis. hepatocytes (TFfl/fl, Albcre) to controls (Table 3). Importantly,
we found a dramatic reduction of TF in TFfl/fl, Albcre mice
Antiphospholipid Syndrome indicating that hepatocytes are the major source of TF in
Antiphospholipid syndrome (APS) is an autoimmune disorder the liver.169 Furthermore, administration of acetaminophen
caused by the development of antibodies against many phos- to wild-type but not TFfl/fl, Albcre mice led to a rapid activa-
pholipids, such as cardiolipin, phospholipid-associated pro- tion of coagulation as measured by increased levels of plasma
teins, including β2-glycoprotein I, and plasma proteins, such thrombin-antithrombin complexes.169 In a second study, we
as prothrombin.241 Collectively, these antibodies are called used a mouse model of bile duct ligation to determine the cel-
antiphospholipid antibodies (aPLA). APS patients with per- lular source of TF responsible for activation of coagulation in
sistently high titers of aPLA are at significant risk of arterial chronic liver injury. As expected, activation of coagulation was
and venous thromboembolic events and fetal loss.242 lower in low TF mice compared with controls.170 Interestingly,
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   719

activation of coagulation was reduced by deletion of TF in Conclusions


hepatocytes but not myeloid cells.170 These 2 mouse models It is clear that the TF-FVIIa complex is essential for hemostasis
indicate that de-encryption of hepatocyte TF mediates the and provides additional protection to vital organs, such as the
rapid activation of coagulation during liver injury. heart, lung, and brain. Further studies are needed to understand
the cell types that express TF in different tissues and control
Sickle Cell Disease hemostasis under basal and pathological conditions. TF con-
SCD is an autosomal recessive disorder caused by a single tributes to arterial and venous thrombosis. TF also plays a
point mutation in the β-globin gene. Under hypoxic condi- major role in the formation of thrombi after rupture of athero-
tions aggregation of the hemoglobin variant leads to the for- sclerotic plaques. Infection induces TF expression on mono-
mation of sickled erythrocytes. Patients with this disorder have cytes and this can trigger DIC. Inhibition of all sources of TF is
an increased risk of both arterial and venous thromboembolic associated with increased bleeding. However, targeted inhibi-
events.257–259 SCD patients have increased numbers of mono- tion of pathological TF expression, for instance in monocytes,
cyte- and EC-derived TF-positive microvesicles and levels may reduce thrombosis associated with different diseases.
were positively correlated with both thrombin-antithrombin
and D-Dimer, which suggested that these particles may con- Acknowledgments
tribute to the procoagulant state observed in SCD.201 We would like to acknowledge Drs D. Monroe, J. Erlich, and Y.
Mouse models have been used to study the role of TF Hisada for helpful comments.
in the activation of coagulation and inflammation in SCD.
Sources of Funding
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Inhibition of TF reduced the chronic procoagulant state and


inflammation observed in SCD mice.73 Furthermore, SCD This work was supported by funding from the John C. Parker
mice had increased thrombosis in a light-dye–mediated injury Professorship.
to the cerebral microcirculation and this was inhibited by an
inhibitory anti-TF antibody.260 Several studies have investi- Disclosures
gated the cellular sources of TF expression that contribute to None.
the activation of coagulation in SCD mice.261 TF expression
has been reported in monocytes, neutrophils, and ECs in SCD References
mice.73,81,82 An early study observed increased TF expression 1. Nemerson Y. Tissue factor and hemostasis. Blood. 1988;71:1–8.
on pulmonary veins of moderate and severe models of SCD 2. Mackman N, Tilley RE, Key NS. Role of the extrinsic pathway of blood
coagulation in hemostasis and thrombosis. Arterioscler Thromb Vasc Biol.
whereas hypoxia and reoxygenation was required to increase 2007;27:1687–1693. doi: 10.1161/ATVBAHA.107.141911.
EC TF expression in a mild model of SCD.81 We found that 3. Edgington TS, Ruf W, Rehemtulla A, Mackman N. The molecular biol-
deletion of TF in ECs did not affect activation of coagulation ogy of initiation of coagulation by tissue factor. Curr Stud Hematol Blood
Transfus. 1991;58:15–21.
in mice with sickle cell bone marrow but reduced levels of
4. Morrissey JH. Tissue factor: an enzyme cofactor and a true receptor.
interleukin6.262 Similar results were observed in SCD mice Thromb Haemost. 2001;86:66–74.
that received the FXa inhibitor rivaroxaban and SCD mice 5. Eilertsen KE, Østerud B. Tissue factor: (patho)physiology and cellular
lacking PAR2 in nonhematopoietic cells.262 These data sug- biology. Blood Coagul Fibrinolysis. 2004;15:521–538.
6. Rao LV, Pendurthi UR. Regulation of tissue factor coagulant activity
gest that a TF-FXa-PAR2 complex on ECs does not contribute on cell surfaces. J Thromb Haemost. 2012;10:2242–2253. doi: 10.1111/
to the activation of coagulation but drives interleukin6 expres- jth.12003.
sion in SCD. 7. Davidson CJ, Tuddenham EG, McVey JH. 450 million years of hemosta-
sis. J Thromb Haemost. 2003;1:1487–1494.
8. Morawitz P. Beitrage zur kenntnis der blutgerinnung. Beitr Chem Physio.
Viral Infections Pathol. 1904;5:133–141.
Viral infections are associated with activation of coagulation 9. Hvatum M, Prydz H. Studies on tissue thromboplastin: I. Solubilization
with sodium deoxycholate. Biochim Biophys Acta. 1966;130:92–101.
and thrombosis. For instance, infection with hemorrhagic 10. Nemerson Y. The phospholipid requirement of tissue factor in blood coag-
fever viruses, such as Ebola virus, is associated with DIC, ulation. J Clin Invest. 1968;47:72–80. doi: 10.1172/JCI105716.
whereas HIV infection increases the risk of thrombosis. 11. Nemerson Y, Pitlick FA. Purification and characterization of the protein
Viruses induce TF expression in a variety of cells, includ- component of tissue factor. Biochemistry. 1970;9:5100–5105.
12. Bjorklid E, Storm E. Purification and some properties of the protein
ing monocytes and ECs.263 Importantly, Ebola virus induced component of tissue thromboplastin from human brain. Biochem J.
TF mRNA expression in peripheral blood mononuclear 1977;165:89–96.
cells in vitro and peripheral blood mononuclear cells iso- 13. Fisher KL, Gorman CM, Vehar GA, O’Brien DP, Lawn RM. Cloning and
expression of human tissue factor cDNA. Thromb Res. 1987;48:89–99.
lated from infected monkeys.202 Moreover, inhibition of TF 14. Morrissey JH, Fakhrai H, Edgington TS. Molecular cloning of the cDNA
reduced mortality in monkeys infected with Ebola virus.264 for tissue factor, the cellular receptor for the initiation of the coagulation
Similarly, monocyte TF expression correlated with levels of protease cascade. Cell. 1987;50:129–135.
HIV and D-dimer in plasma from HIV infected patients.203 15. Scarpati EM, Wen D, Broze GJ Jr, Miletich JP, Flandermeyer RR, Siegel
NR, Sadler JE. Human tissue factor: cDNA sequence and chromosome
More recently, it was shown that TF is expressed by a sub- localization of the gene. Biochemistry. 1987;26:5234–5238.
set of monocytes (CD14+, CD16−, CCR2+) in HIV infected 16. Spicer EK, Horton R, Bloem L, Bach R, Williams KR, Guha A, Kraus J,
patients, and inhibition of TF reduced D-dimer levels in mon- Lin TC, Nemerson Y, Konigsberg WH. Isolation of cDNA clones coding
for human tissue factor: primary structure of the protein and cDNA. Proc
keys infected with simian immunodeficiency virus.106 These
Natl Acad Sci USA. 1987;84:5148–5152.
studies suggest that TF expression by monocyte drives the 17. Mackman N, Morrissey JH, Fowler B, Edgington TS. Complete

coagulopathy in HIV infected patients. sequence of the human tissue factor gene, a highly regulated cellular
720   Arterioscler Thromb Vasc Biol   April 2018

receptor that initiates the coagulation protease cascade. Biochemistry. 40. Morrissey J, Drake T, Schlag G, Redl H. Procoagulant Response of the
1989;28:1755–1762. Endothelium and Monocytes. Berlin, New York: Springer Verlag; 1993.
18. Harlos K, Martin DM, O’Brien DP, Jones EY, Stuart DI, Polikarpov I, 41. Parry GC, Mackman N. Transcriptional regulation of tissue factor

Miller A, Tuddenham EG, Boys CW. Crystal structure of the extra- expression in human endothelial cells. Arterioscler Thromb Vasc Biol.
cellular region of human tissue factor. Nature. 1994;370:662–666. doi: 1995;15:612–621.
10.1038/370662a0. 42. Schwertz H, Tolley ND, Foulks JM, Denis MM, Risenmay BW, Buerke
19. Bugge TH, Xiao Q, Kombrinck KW, Flick MJ, Holmbäck K, Danton MJ, M, Tilley RE, Rondina MT, Harris EM, Kraiss LW, Mackman N,
Colbert MC, Witte DP, Fujikawa K, Davie EW, Degen JL. Fatal embryonic Zimmerman GA, Weyrich AS. Signal-dependent splicing of tissue factor
bleeding events in mice lacking tissue factor, the cell-associated initiator pre-mRNA modulates the thrombogenicity of human platelets. J Exp Med.
of blood coagulation. Proc Natl Acad Sci USA. 1996;93:6258–6263. 2006;203:2433–2440. doi: 10.1084/jem.20061302.
20. Carmeliet P, Mackman N, Moons L, Luther T, Gressens P, Van Vlaenderen 43. Camera M, Frigerio M, Toschi V, Brambilla M, Rossi F, Cottell DC,
I, Demunck H, Kasper M, Breier G, Evrard P, Müller M, Risau W, Maderna P, Parolari A, Bonzi R, De Vincenti O, Tremoli E. Platelet activa-
Edgington T, Collen D. Role of tissue factor in embryonic blood vessel tion induces cell-surface immunoreactive tissue factor expression, which
development. Nature. 1996;383:73–75. doi: 10.1038/383073a0. is modulated differently by antiplatelet drugs. Arterioscler Thromb Vasc
21. Toomey JR, Kratzer KE, Lasky NM, Stanton JJ, Broze GJ Jr. Targeted Biol. 2003;23:1690–1696. doi: 10.1161/01.ATV.0000085629.23209.AA.
disruption of the murine tissue factor gene results in embryonic lethality. 44. Panes O, Matus V, Sáez CG, Quiroga T, Pereira J, Mezzano D. Human
Blood. 1996;88:1583–1587. platelets synthesize and express functional tissue factor. Blood.
22. Hoffman M, Monroe DM III. A cell-based model of hemostasis. Thromb 2007;109:5242–5250. doi: 10.1182/blood-2006-06-030619.
Haemost. 2001;85:958–965. 45. Brambilla M, Rossetti L, Zara C, Canzano P, Giesen PLA, Tremoli E,
23. Yamamoto M, Nakagaki T, Kisiel W. Tissue factor-dependent autoactivation Camera M. Do methodological differences account for the current contro-
of human blood coagulation factor VII. J Biol Chem. 1992;267:19089–19094. versy on tissue factor expression in platelets? Platelets. 2017:1–9.
24. Josso F, Prou-Wartelle O. Interaction of tissue factor and factor VII at the ear- 46. Brambilla M, Facchinetti L, Canzano P, Rossetti L, Ferri N, Balduini A,
liest phase of coagulation. Thromb Diath Haemorrh Suppl. 1965;17:35–44. Abbonante V, Boselli D, De Marco L, Di Minno MN, Toschi V, Corsini
25. Osterud B, Rapaport SI. Activation of factor IX by the reaction product of A, Tremoli E, Camera M. Human megakaryocytes confer tissue factor
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

tissue factor and factor VII: additional pathway for initiating blood coagu- to a subset of shed platelets to stimulate thrombin generation. Thromb
lation. Proc Natl Acad Sci USA. 1977;74:5260–5264. Haemost. 2015;114:579–592. doi: 10.1160/TH14-10-0830.
26. Zur M, Nemerson Y. Kinetics of factor IX activation via the extrin- 47. Gerrits AJ, Koekman CA, van Haeften TW, Akkerman JW. Platelet tis-
sic pathway. Dependence of Km on tissue factor. J Biol Chem. sue factor synthesis in type 2 diabetic patients is resistant to inhibition by
1980;255:5703–5707. insulin. Diabetes. 2010;59:1487–1495. doi: 10.2337/db09-1008.
27. Lu G, Broze GJ Jr, Krishnaswamy S. Formation of factors IXa and Xa 48. Brambilla M, Camera M, Colnago D, Marenzi G, De Metrio M, Giesen
by the extrinsic pathway: differential regulation by tissue factor pathway PL, Balduini A, Veglia F, Gertow K, Biglioli P, Tremoli E. Tissue factor
inhibitor and antithrombin III. J Biol Chem. 2004;279:17241–17249. doi: in patients with acute coronary syndromes: expression in platelets, leuko-
10.1074/jbc.M312827200. cytes, and platelet-leukocyte aggregates. Arterioscler Thromb Vasc Biol.
28. Taylor SA, Liddell MB, Peake IR, Bloom AL, Lillicrap DP. A mutation 2008;28:947–953. doi: 10.1161/ATVBAHA.107.161471.
adjacent to the beta cleavage site of factor IX (valine 182 to leucine) 49. Østerud B, Olsen JO. Human platelets do not express tissue factor. Thromb
results in mild haemophilia Bm. Br J Haematol. 1990;75:217–221. Res. 2013;132:112–115. doi: 10.1016/j.thromres.2013.04.010.
29. Kamikubo Y, Mendolicchio GL, Zampolli A, Marchese P, Rothmeier AS, 50. Bouchard BA, Mann KG, Butenas S. No evidence for tissue factor on
Orje JN, Gale AJ, Krishnaswamy S, Gruber A, Østergaard H, Petersen platelets. Blood. 2010;116:854–855. doi: 10.1182/blood-2010-05-285627.
LC, Ruf W, Ruggeri ZM. Selective factor VIII activation by the tissue 51. Bouchard BA, Krudysz-Amblo J, Butenas S. Platelet tissue factor is not
factor-factor VIIa-factor Xa complex. Blood. 2017;130:1661–1670. doi: expressed transiently after platelet activation. Blood. 2012;119:4338–
10.1182/blood-2017-02-767079. 4339; author reply 4339. doi: 10.1182/blood-2012-01-403469.
30. Bouchard BA, Chapin J, Brummel-Ziedins KE, Durda P, Key NS, Tracy 52. Ritis K, Doumas M, Mastellos D, Micheli A, Giaglis S, Magotti P, Rafail
PB. Platelets and platelet-derived factor Va confer hemostatic compe- S, Kartalis G, Sideras P, Lambris JD. A novel C5a receptor-tissue factor
tence in complete factor V deficiency. Blood. 2015;125:3647–3650. doi: cross-talk in neutrophils links innate immunity to coagulation pathways. J
10.1182/blood-2014-07-589580. Immunol. 2006;177:4794–4802.
31. Gailani D, Broze GJ Jr. Factor XI activation in a revised model of blood 53. Kambas K, Markiewski MM, Pneumatikos IA, Rafail SS, Theodorou V,
coagulation. Science. 1991;253:909–912. Konstantonis D, Kourtzelis I, Doumas MN, Magotti P, Deangelis RA,
32. Giesen PL, Rauch U, Bohrmann B, Kling D, Roqué M, Fallon JT,
Lambris JD, Ritis KD. C5a and TNF-alpha up-regulate the expression of
Badimon JJ, Himber J, Riederer MA, Nemerson Y. Blood-borne tissue tissue factor in intra-alveolar neutrophils of patients with the acute respira-
factor: another view of thrombosis. Proc Natl Acad Sci USA. 1999;96: tory distress syndrome. J Immunol. 2008;180:7368–7375.
2311–2315. 54. Osterud B, Rao LV, Olsen JO. Induction of tissue factor expression in
33. Butenas S, Mann KG. Active tissue factor in blood? Nat Med.
whole blood: lack of evidence for the presence of tissue factor expression
2004;10:1155–1156; author reply 1156. doi: 10.1038/nm1104-1155b. in granulocytes. Thromb Haemost. 2000;83:861–867.
34. Butenas S, Bouchard BA, Brummel-Ziedins KE, Parhami-Seren B, Mann 55. Moosbauer C, Morgenstern E, Cuvelier SL, Manukyan D, Bidzhekov K,
KG. Tissue factor activity in whole blood. Blood. 2005;105:2764–2770. Albrecht S, Lohse P, Patel KD, Engelmann B. Eosinophils are a major
doi: 10.1182/blood-2004-09-3567. intravascular location for tissue factor storage and exposure. Blood.
35. Hoffman M, Whinna HC, Monroe DM. Circulating tissue factor accu- 2007;109:995–1002. doi: 10.1182/blood-2006-02-004945.
mulates in thrombi, but not in hemostatic plugs. J Thromb Haemost. 56. Uderhardt S, Ackermann JA, Fillep T, et al. Enzymatic lipid oxidation by
2006;4:2092–2093. doi: 10.1111/j.1538-7836.2006.02085.x. eosinophils propagates coagulation, hemostasis, and thrombotic disease.
36. Egorina EM, Sovershaev MA, Bjørkøy G, Gruber FX, Olsen JO,
J Exp Med. 2017;214:2121–2138. doi: 10.1084/jem.20161070.
Parhami-Seren B, Mann KG, Østerud B. Intracellular and surface distri- 57. Sovershaev MA, Lind KF, Devold H, Jørgensen TØ, Hansen JB,

bution of monocyte tissue factor: application to intersubject variability. Østerud B, Egorina EM. No evidence for the presence of tissue factor
Arterioscler Thromb Vasc Biol. 2005;25:1493–1498. doi: 10.1161/01. in high-purity preparations of immunologically isolated eosinophils. J
ATV.0000168413.29874.d7. Thromb Haemost. 2008;6:1742–1749. doi: 10.1111/j.1538-7836.2008.
37. Gregory SA, Morrissey JH, Edgington TS. Regulation of tissue factor 03105.x.
gene expression in the monocyte procoagulant response to endotoxin. Mol 58. De Palma R, Cirillo P, Ciccarelli G, Barra G, Conte S, Pellegrino G,
Cell Biol. 1989;9:2752–2755. Pasquale G, Nassa G, Pacifico F, Leonardi A, Insabato L, Calì G,
38. Drake TA, Cheng J, Chang A, Taylor FB Jr. Expression of tissue factor, Golino P, Cimmino G. Expression of functional tissue factor in activated
thrombomodulin, and E-selectin in baboons with lethal Escherichia coli T-lymphocytes in vitro and in vivo: A possible contribution of immu-
sepsis. Am J Pathol. 1993;142:1458–1470. nity to thrombosis? Int J Cardiol. 2016;218:188–195. doi: 10.1016/j.
39. Franco RF, de Jonge E, Dekkers PE, Timmerman JJ, Spek CA, van ijcard.2016.04.177.
Deventer SJ, van Deursen P, van Kerkhoff L, van Gemen B, ten Cate H, 59. Mackman N, Brand K, Edgington TS. Lipopolysaccharide-mediated tran-
van der Poll T, Reitsma PH. The in vivo kinetics of tissue factor messenger scriptional activation of the human tissue factor gene in THP-1 monocytic
RNA expression during human endotoxemia: relationship with activation cells requires both activator protein 1 and nuclear factor kappa B binding
of coagulation. Blood. 2000;96:554–559. sites. J Exp Med. 1991;174:1517–1526.
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   721

60. Osterud B. Tissue factor expression by monocytes: regulation and patho- 83. Song D, Ye X, Xu H, Liu SF. Activation of endothelial intrinsic NF-
physiological roles. Blood Coagul Fibrinolysis. 1998;9(suppl 1):S9–S14. {kappa}B pathway impairs protein C anticoagulation mechanism and
61. Mackman N. Regulation of the tissue factor gene. FASEB J. 1995;9: promotes coagulation in endotoxemic mice. Blood. 2009;114:2521–
883–889. 2529. doi: 10.1182/blood-2009-02-205914.
62. Osterud B, Bjorklid E. Tissue factor in blood cells and endothelial cells. 84. Fleck RA, Rao LV, Rapaport SI, Varki N. Localization of human tissue
Front Biosci (Elite Ed). 2012;4:289–299. factor antigen by immunostaining with monospecific, polyclonal anti-
63. Rauch U, Bonderman D, Bohrmann B, Badimon JJ, Himber J, Riederer human tissue factor antibody. Thromb Res. 1990;59:421–437.
MA, Nemerson Y. Transfer of tissue factor from leukocytes to platelets is 85. Bach RR. Tissue factor encryption. Arterioscler Thromb Vasc Biol.
mediated by CD15 and tissue factor. Blood. 2000;96:170–175. 2006;26:456–461. doi: 10.1161/01.ATV.0000202656.53964.04.
64. Del Conde I, Shrimpton CN, Thiagarajan P, López JA. Tissue-factor-bearing 86. Bach R, Rifkin DB. Expression of tissue factor procoagulant ac-
microvesicles arise from lipid rafts and fuse with activated platelets to initiate tivity: regulation by cytosolic calcium. Proc Natl Acad Sci USA.
coagulation. Blood. 2005;106:1604–1611. doi: 10.1182/blood-2004-03-1095. 1990;87:6995–6999.
65. Sovershaev MA, Egorina EM, Osterud B, Hansen JB. Evidence for direct trans- 87. Le DT, Rapaport SI, Rao LV. Relations between factor VIIa binding and
fer of tissue factor from monocytes to platelets in whole blood. Blood Coagul expression of factor VIIa/tissue factor catalytic activity on cell surfaces.
Fibrinolysis. 2012;23:345–350. doi: 10.1097/MBC.0b013e328350bf2f. J Biol Chem. 1992;267:15447–15454.
66. Egorina EM, Sovershaev MA, Olsen JO, Østerud B. Granulocytes do 88. Bach RR, Moldow CF. Mechanism of tissue factor activation on HL-60
not express but acquire monocyte-derived tissue factor in whole blood: cells. Blood. 1997;89:3270–3276.
evidence for a direct transfer. Blood. 2008;111:1208–1216. doi: 10.1182/ 89. Bach R, Gentry R, Nemerson Y. Factor VII binding to tissue factor in
blood-2007-08-107698. reconstituted phospholipid vesicles: induction of cooperativity by phos-
67. Camera M, Brambilla M, Facchinetti L, Canzano P, Spirito R, Rossetti L, phatidylserine. Biochemistry. 1986;25:4007–4020.
Saccu C, Di Minno MN, Tremoli E. Tissue factor and atherosclerosis: not 90. Ohkubo YZ, Morrissey JH, Tajkhorshid E. Dynamical view of
only vessel wall-derived TF, but also platelet-associated TF. Thromb Res. membrane binding and complex formation of human factor VIIa
2012;129:279–284. doi: 10.1016/j.thromres.2011.11.028. and tissue factor. J Thromb Haemost. 2010;8:1044–1053. doi:
68. Wang JG, Geddings JE, Aleman MM, et al. Tumor-derived tissue fac- 10.1111/j.1538-7836.2010.03826.x.
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

tor activates coagulation and enhances thrombosis in a mouse xenograft 91. Shaw AW, Pureza VS, Sligar SG, Morrissey JH. The local phospholipid
model of human pancreatic cancer. Blood. 2012;119:5543–5552. doi: environment modulates the activation of blood clotting. J Biol Chem.
10.1182/blood-2012-01-402156. 2007;282:6556–6563. doi: 10.1074/jbc.M607973200.
69. Mackman N, Luther T. Platelet tissue factor: to be or not to be. Thromb 92. Tavoosi N, Davis-Harrison RL, Pogorelov TV, Ohkubo YZ, Arcario
Res. 2013;132:3–5. doi: 10.1016/j.thromres.2013.05.009. MJ, Clay MC, Rienstra CM, Tajkhorshid E, Morrissey JH. Molecular
70. Todoroki H, Higure A, Okamoto K, Okazaki K, Nagafuchi Y, Takeda S, determinants of phospholipid synergy in blood clotting. J Biol Chem.
Katoh H, Itoh H, Ohsato K, Nakamura S. Possible role of platelet-activat- 2011;286:23247–23253. doi: 10.1074/jbc.M111.251769.
ing factor in the in vivo expression of tissue factor in neutrophils. J Surg 93. Wang J, Pendurthi UR, Rao LVM. Sphingomyelin encrypts tissue fac-
Res. 1998;80:149–155. doi: 10.1006/jsre.1998.5348. tor: ATP-induced activation of A-SMase leads to tissue factor decryption
71. Todoroki H, Nakamura S, Higure A, Okamoto K, Takeda S, Nagata N, and microvesicle shedding. Blood Adv. 2017;1:849–862. doi: 10.1182/
Itoh H, Ohsato K. Neutrophils express tissue factor in a monkey model of bloodadvances.2016003947.
sepsis. Surgery. 2000;127:209–216. doi: 10.1067/msy.2000.103027. 94. Rehemtulla A, Ruf W, Edgington TS. The integrity of the cysteine
72. Darbousset R, Thomas GM, Mezouar S, Frère C, Bonier R, Mackman N, 186-cysteine 209 bond of the second disulfide loop of tissue factor is
Renné T, Dignat-George F, Dubois C, Panicot-Dubois L. Tissue factor-pos- required for binding of factor VII. J Biol Chem. 1991;266:10294–10299.
itive neutrophils bind to injured endothelial wall and initiate thrombus forma- 95. Ahamed J, Versteeg HH, Kerver M, Chen VM, Mueller BM, Hogg PJ,
tion. Blood. 2012;120:2133–2143. doi: 10.1182/blood-2012-06-437772. Ruf W. Disulfide isomerization switches tissue factor from coagulation
73. Chantrathammachart P, Mackman N, Sparkenbaugh E, Wang JG, Parise to cell signaling. Proc Natl Acad Sci USA. 2006;103:13932–13937. doi:
LV, Kirchhofer D, Key NS, Pawlinski R. Tissue factor promotes activation 10.1073/pnas.0606411103.
of coagulation and inflammation in a mouse model of sickle cell disease. 96. Versteeg HH, Ruf W. Tissue factor coagulant function is enhanced by
Blood. 2012;120:636–646. doi: 10.1182/blood-2012-04-424143. protein-disulfide isomerase independent of oxidoreductase activity. J
74. Antoniak S, Mackman N. Editorial commentary: tissue factor expression Biol Chem. 2007;282:25416–25424. doi: 10.1074/jbc.M702410200.
by the endothelium: coagulation or inflammation? Trends Cardiovasc 97. Reinhardt C, von Brühl ML, Manukyan D, Grahl L, Lorenz M, Altmann
Med. 2016;26:304–305. doi: 10.1016/j.tcm.2015.12.002. B, Dlugai S, Hess S, Konrad I, Orschiedt L, Mackman N, Ruddock L,
75. Witkowski M, Landmesser U, Rauch U. Tissue factor as a link between Massberg S, Engelmann B. Protein disulfide isomerase acts as an injury
inflammation and coagulation. Trends Cardiovasc Med. 2016;26:297– response signal that enhances fibrin generation via tissue factor activa-
303. doi: 10.1016/j.tcm.2015.12.001. tion. J Clin Invest. 2008;118:1110–1122. doi: 10.1172/JCI32376.
76. Antoniak S, Mackman N. Letter to editor response: endothelial cell tissue 98. Langer F, Spath B, Fischer C, Stolz M, Ayuk FA, Kröger N, Bokemeyer
factor and coagulation. Trends Cardiovasc Med. 2017;27:157. C, Ruf W. Rapid activation of monocyte tissue factor by antithymo-
77. Crossman DC, Carr DP, Tuddenham EG, Pearson JD, McVey JH. The cyte globulin is dependent on complement and protein disulfide
regulation of tissue factor mRNA in human endothelial cells in response isomerase. Blood. 2013;121:2324–2335. doi: 10.1182/blood-2012-
to endotoxin or phorbol ester. J Biol Chem. 1990;265:9782–9787. 10-460493.
78. Bevilacqua MP, Pober JS, Majeau GR, Cotran RS, Gimbrone MA Jr. 99. Jasuja R, Passam FH, Kennedy DR, Kim SH, van Hessem L, Lin L,
Interleukin 1 (IL-1) induces biosynthesis and cell surface expression of Bowley SR, Joshi SS, Dilks JR, Furie B, Furie BC, Flaumenhaft R.
procoagulant activity in human vascular endothelial cells. J Exp Med. Protein disulfide isomerase inhibitors constitute a new class of anti-
1984;160:618–623. thrombotic agents. J Clin Invest. 2012;122:2104–2113. doi: 10.1172/
79. Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of JCI61228.
tissue factor in human tissues. Implications for disorders of hemostasis 100. Bach RR, Monroe D. What is wrong with the allosteric disulfide bond
and thrombosis. Am J Pathol. 1989;134:1087–1097. hypothesis? Arterioscler Thromb Vasc Biol. 2009;29:1997–1998. doi:
80. Lupu C, Westmuckett AD, Peer G, Ivanciu L, Zhu H, Taylor FB Jr, Lupu F. 10.1161/ATVBAHA.109.194985.
Tissue factor-dependent coagulation is preferentially up-regulated within 101. Popescu NI, Lupu C, Lupu F. Extracellular protein disulfide isomerase
arterial branching areas in a baboon model of Escherichia coli sepsis. Am regulates coagulation on endothelial cells through modulation of phos-
J Pathol. 2005;167:1161–1172. doi: 10.1016/S0002-9440(10)61204-7. phatidylserine exposure. Blood. 2010;116:993–1001. doi: 10.1182/
81. Solovey A, Kollander R, Shet A, Milbauer LC, Choong S, Panoskaltsis- blood-2009-10-249607.
Mortari A, Blazar BR, Kelm RJ Jr, Hebbel RP. Endothelial cell expression 102. Golino P. The inhibitors of the tissue factor:factor VII pathway. Thromb
of tissue factor in sickle mice is augmented by hypoxia/reoxygenation Res. 2002;106:V257–V265.
and inhibited by lovastatin. Blood. 2004;104:840–846. doi: 10.1182/ 103. Lee AY, Vlasuk GP. Recombinant nematode anticoagulant protein c2 and
blood-2003-10-3719. other inhibitors targeting blood coagulation factor VIIa/tissue factor. J
82. Solovey A, Kollander R, Milbauer LC, Abdulla F, Chen Y, Kelm RJ Jr, Intern Med. 2003;254:313–321.
Hebbel RP. Endothelial nitric oxide synthase and nitric oxide regulate 104. Francischetti IM, Valenzuela JG, Andersen JF, Mather TN, Ribeiro JM.
endothelial tissue factor expression in vivo in the sickle transgenic mouse. Ixolaris, a novel recombinant tissue factor pathway inhibitor (TFPI)
Am J Hematol. 2010;85:41–45. doi: 10.1002/ajh.21582. from the salivary gland of the tick, Ixodes scapularis: identification of
722   Arterioscler Thromb Vasc Biol   April 2018

factor X and factor Xa as scaffolds for the inhibition of factor VIIa/ ligation. Proc Natl Acad Sci USA. 2009;106:19497–19502. doi: 10.1073/
tissue factor complex. Blood. 2002;99:3602–3612. doi: 10.1182/ pnas.0905325106.
blood-2001-12-0237. 125. Kocatürk B, Van den Berg YW, Tieken C, Mieog JS, de Kruijf EM, Engels
105. Giugliano RP, Wiviott SD, Stone PH, Simon DI, Schweiger MJ,
CC, van der Ent MA, Kuppen PJ, Van de Velde CJ, Ruf W, Reitsma PH,
Bouchard A, Leesar MA, Goulder MA, Deitcher SR, McCabe CH, Osanto S, Liefers GJ, Bogdanov VY, Versteeg HH. Alternatively spliced
Braunwald E; ANTHEM-TIMI-32 Investigators. Recombinant nematode tissue factor promotes breast cancer growth in a β1 integrin-dependent
anticoagulant protein c2 in patients with non-ST-segment elevation acute manner. Proc Natl Acad Sci USA. 2013;110:11517–11522. doi: 10.1073/
coronary syndrome: the ANTHEM-TIMI-32 trial. J Am Coll Cardiol. pnas.1307100110.
2007;49:2398–2407. doi: 10.1016/j.jacc.2007.02.065. 126. Rosen ED, Chan JC, Idusogie E, Clotman F, Vlasuk G, Luther T, Jalbert
106. Schechter ME, Andrade BB, He T et al. Inflammatory monocytes ex- LR, Albrecht S, Zhong L, Lissens A, Schoonjans L, Moons L, Collen D,
pressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med. Castellino FJ, Carmeliet P. Mice lacking factor VII develop normally but
2017;9:1–13. suffer fatal perinatal bleeding. Nature. 1997;390:290–294.
107. Creasey AA, Chang AC, Feigen L, Wün TC, Taylor FB Jr, Hinshaw 127. Cui J, O’Shea KS, Purkayastha A, Saunders TL, Ginsburg D. Fatal haem-
LB. Tissue factor pathway inhibitor reduces mortality from Escherichia orrhage and incomplete block to embryogenesis in mice lacking coagula-
coli septic shock. J Clin Invest. 1993;91:2850–2860. doi: 10.1172/ tion factor V. Nature. 1996;384:66–68. doi: 10.1038/384066a0.
JCI116529. 128. Dewerchin M, Liang Z, Moons L, Carmeliet P, Castellino FJ, Collen
108. Abraham E, Reinhart K, Opal S, et al; OPTIMIST Trial Study Group. D, Rosen ED. Blood coagulation factor X deficiency causes partial em-
Efficacy and safety of tifacogin (recombinant tissue factor path- bryonic lethality and fatal neonatal bleeding in mice. Thromb Haemost.
way inhibitor) in severe sepsis: a randomized controlled trial. JAMA. 2000;83:185–190.
2003;290:238–247. doi: 10.1001/jama.290.2.238. 129. Sun WY, Witte DP, Degen JL, Colbert MC, Burkart MC, Holmbäck
109. Welty-Wolf KE, Carraway MS, Ortel TL, Ghio AJ, Idell S, Egan J, Zhu K, Xiao Q, Bugge TH, Degen SJ. Prothrombin deficiency results in
X, Jiao JA, Wong HC, Piantadosi CA. Blockade of tissue factor-factor embryonic and neonatal lethality in mice. Proc Natl Acad Sci USA.
X binding attenuates sepsis-induced respiratory and renal failure. Am 1998;95:7597–7602.
J Physiol Lung Cell Mol Physiol. 2006;290:L21–L31. doi: 10.1152/ 130. Xue J, Wu Q, Westfield LA, Tuley EA, Lu D, Zhang Q, Shim K, Zheng
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

ajplung.00155.2005. X, Sadler JE. Incomplete embryonic lethality and fatal neonatal hem-
110. Vincent JL, Artigas A, Petersen LC, Meyer C. A multicenter, randomized, orrhage caused by prothrombin deficiency in mice. Proc Natl Acad Sci
double-blind, placebo-controlled, dose-escalation trial assessing safety USA. 1998;95:7603–7607.
and efficacy of active site inactivated recombinant factor VIIa in subjects 131. Kundu RK, Sangiorgi F, Wu LY, Kurachi K, Anderson WF, Maxson R,
with acute lung injury or acute respiratory distress syndrome. Crit Care Gordon EM. Targeted inactivation of the coagulation factor IX gene
Med. 2009;37:1874–1880. doi: 10.1097/CCM.0b013e31819fff2c. causes hemophilia B in mice. Blood. 1998;92:168–174.
111. Abraham E. Anticoagulants for acute lung injury: barking up the
132. Bi L, Lawler AM, Antonarakis SE, High KA, Gearhart JD, Kazazian HH
wrong tree. Crit Care Med. 2009;37:2100–2101. doi: 10.1097/CCM. Jr. Targeted disruption of the mouse factor VIII gene produces a model of
0b013e3181a5ea63. haemophilia A. Nat Genet. 1995;10:119–121. doi: 10.1038/ng0595-119.
112. Pendurthi UR, Rao LV. Role of tissue factor disulfides and lipid rafts 133. Lin HF, Maeda N, Smithies O, Straight DL, Stafford DW. A coagula-
in signaling. Thromb Res. 2008;122(suppl 1):S14–S18. doi: 10.1016/ tion factor IX-deficient mouse model for human hemophilia B. Blood.
S0049-3848(08)70012-4. 1997;90:3962–3966.
113. Versteeg HH, Schaffner F, Kerver M, Petersen HH, Ahamed J, Felding- 134. Wang L, Zoppè M, Hackeng TM, Griffin JH, Lee KF, Verma IM. A fac-
Habermann B, Takada Y, Mueller BM, Ruf W. Inhibition of tissue fac- tor IX-deficient mouse model for hemophilia B gene therapy. Proc Natl
tor signaling suppresses tumor growth. Blood. 2008;111:190–199. doi: Acad Sci USA. 1997;94:11563–11566.
10.1182/blood-2007-07-101048. 135. Ay C, Hisada Y, Cooley BC, Mackman N. Factor XI-deficient mice ex-
114. Rothmeier AS, Liu E, Chakrabarty S, Disse J, Mueller BM, Østergaard H, hibit increased bleeding after injury to the saphenous vein. J Thromb
Ruf W. Identification of the coagulation factor VIIa integrin binding site Haemost. 2017;15:1829–1833. doi: 10.1111/jth.13766.
required for pro-angiogenic PAR2 signaling [published online ahead of 136. Gailani D, Lasky NM, Broze GJ Jr. A murine model of factor XI defi-
print December 15, 2017]. Blood. doi: 10.1182/blood-2017-02-768218. ciency. Blood Coagul Fibrinolysis. 1997;8:134–144.
115. Coughlin SR. Thrombin signalling and protease-activated receptors.
137. Pauer HU, Renné T, Hemmerlein B, Legler T, Fritzlar S, Adham I,
Nature. 2000;407:258–264. doi: 10.1038/35025229. Müller-Esterl W, Emons G, Sancken U, Engel W, Burfeind P. Targeted
116. Riewald M, Ruf W. Mechanistic coupling of protease signaling and deletion of murine coagulation factor XII gene-a model for contact phase
initiation of coagulation by tissue factor. Proc Natl Acad Sci USA. activation in vivo. Thromb Haemost. 2004;92:503–508. doi: 10.1160/
2001;98:7742–7747. doi: 10.1073/pnas.141126698. TH04-04-0250.
117. Schaffner F, Ruf W. Tissue factor and PAR2 signaling in the tumor mi- 138. Renné T, Pozgajová M, Grüner S, Schuh K, Pauer HU, Burfeind P,
croenvironment. Arterioscler Thromb Vasc Biol. 2009;29:1999–2004. Gailani D, Nieswandt B. Defective thrombus formation in mice lack-
doi: 10.1161/ATVBAHA.108.177428. ing coagulation factor XII. J Exp Med. 2005;202:271–281. doi: 10.1084/
118. Bogdanov VY, Balasubramanian V, Hathcock J, Vele O, Lieb M,
jem.20050664.
Nemerson Y. Alternatively spliced human tissue factor: a circulating, 139. Kleinschnitz C, Stoll G, Bendszus M, Schuh K, Pauer HU, Burfeind
soluble, thrombogenic protein. Nat Med. 2003;9:458–462. doi: 10.1038/ P, Renné C, Gailani D, Nieswandt B, Renné T. Targeting coagulation
nm841. factor XII provides protection from pathological thrombosis in cerebral
119. Bogdanov VY, Kirk RI, Miller C, Hathcock JJ, Vele S, Gazdoiu M, ischemia without interfering with hemostasis. J Exp Med. 2006;203:513–
Nemerson Y, Taubman MB. Identification and characterization of murine 518. doi: 10.1084/jem.20052458.
alternatively spliced tissue factor. J Thromb Haemost. 2006;4:158–167. 140. Acharya SS, Coughlin A, Dimichele DM; North American Rare

doi: 10.1111/j.1538-7836.2005.01680.x. Bleeding Disorder Study Group. Rare bleeding disorder registry: defi-
120. Mackman N. Alternatively spliced tissue factor - one cut too many? ciencies of factors II, V, VII, X, XIII, fibrinogen and dysfibrinogenemias.
Thromb Haemost. 2007;97:5–8. J Thromb Haemost. 2004;2:248–256. doi: 10.1111/j.1538-7836.2003.
121. Censarek P, Bobbe A, Grandoch M, Schrör K, Weber AA. Alternatively t01-1-00553.x.
spliced human tissue factor (asHTF) is not pro-coagulant. Thromb 141. Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet.

Haemost. 2007;97:11–14. 2003;361:1801–1809. doi: 10.1016/S0140-6736(03)13405-8.
122. Ruf W, Miles DJ, Rehemtulla A, Edgington TS. Tissue factor residues 142. Duga S, Salomon O. Congenital factor XI deficiency: an update. Semin
157-167 are required for efficient proteolytic activation of factor X and Thromb Hemost. 2013;39:621–631. doi: 10.1055/s-0033-1353420.
factor VII. J Biol Chem. 1992;267:22206–22210. 143. Salomon O, Steinberg DM, Seligshon U. Variable bleeding manifes-
123. Szotowski B, Antoniak S, Poller W, Schultheiss HP, Rauch U.
tations characterize different types of surgery in patients with severe
Procoagulant soluble tissue factor is released from endothelial cells in factor XI deficiency enabling parsimonious use of replacement thera-
response to inflammatory cytokines. Circ Res. 2005;96:1233–1239. doi: py. Haemophilia. 2006;12:490–493. doi: 10.1111/j.1365-2516.2006.
10.1161/01.RES.0000171805.24799.fa. 01304.x.
124. van den Berg YW, van den Hengel LG, Myers HR, Ayachi O, Jordanova 144. Bolton-Maggs PH, Young Wan-Yin B, McCraw AH, Slack J, Kernoff
E, Ruf W, Spek CA, Reitsma PH, Bogdanov VY, Versteeg HH. PB. Inheritance and bleeding in factor XI deficiency. Br J Haematol.
Alternatively spliced tissue factor induces angiogenesis through integrin 1988;69:521–528.
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   723

145. Bouchard BA, Shatos MA, Tracy PB. Human brain pericytes differen- 165. Petersen LC, Nørby PL, Branner S, Sørensen BB, Elm T, Stennicke
tially regulate expression of procoagulant enzyme complexes comprising HR, Persson E, Bjørn SE. Characterization of recombinant murine fac-
the extrinsic pathway of blood coagulation. Arterioscler Thromb Vasc tor VIIa and recombinant murine tissue factor: a human-murine spe-
Biol. 1997;17:1–9. cies compatibility study. Thromb Res. 2005;116:75–85. doi: 10.1016/j.
146. Hoffman M, Colina CM, McDonald AG, Arepally GM, Pedersen L, thromres.2004.11.003.
Monroe DM. Tissue factor around dermal vessels has bound factor VII 166. Wang JG, Manly D, Kirchhofer D, Pawlinski R, Mackman N. Levels
in the absence of injury. J Thromb Haemost. 2007;5:1403–1408. doi: of microparticle tissue factor activity correlate with coagulation activa-
10.1111/j.1538-7836.2007.02576.x. tion in endotoxemic mice. J Thromb Haemost. 2009;7:1092–1098. doi:
147. Mackman N. The role of tissue factor and factor VIIa in hemostasis. Anesth 10.1111/j.1538-7836.2009.03448.x.
Analg. 2009;108:1447–1452. doi: 10.1213/ane.0b013e31819bceb1. 167. Wang L, Miller C, Swarthout RF, Rao M, Mackman N, Taubman MB.
148. Miller GJ, Howarth DJ, Attfield JC, Cooke CJ, Nanjee MN, Olszewski Vascular smooth muscle-derived tissue factor is critical for arterial
WL, Morrissey JH, Miller NE. Haemostatic factors in human peripheral thrombosis after ferric chloride-induced injury. Blood. 2009;113:705–
afferent lymph. Thromb Haemost. 2000;83:427–432. 713. doi: 10.1182/blood-2007-05-090944.
149. Hoffman M, Monroe DM. Tissue factor in brain is not saturated with 168. Pawlinski R, Wang JG, Owens AP III, Williams J, Antoniak S, Tencati M,
factor VIIa: implications for factor VIIa dosing in intracerebral hem- Luther T, Rowley JW, Low EN, Weyrich AS, Mackman N. Hematopoietic
orrhage. Stroke. 2009;40:2882–2884. doi: 10.1161/STROKEAHA. and nonhematopoietic cell tissue factor activates the coagulation cas-
109.555433. cade in endotoxemic mice. Blood. 2010;116:806–814. doi: 10.1182/
150. Parry GC, Erlich JH, Carmeliet P, Luther T, Mackman N. Low levels of blood-2009-12-259267.
tissue factor are compatible with development and hemostasis in mice. J 169. Sullivan BP, Kopec AK, Joshi N, Cline H, Brown JA, Bishop SC, Kassel
Clin Invest. 1998;101:560–569. doi: 10.1172/JCI814. KM, Rockwell C, Mackman N, Luyendyk JP. Hepatocyte tissue factor
151. Rosen ED, Xu H, Liang Z, Martin JA, Suckow M, Castellino FJ.
activates the coagulation cascade in mice. Blood. 2013;121:1868–1874.
Generation of genetically-altered mice producing very low levels of co- doi: 10.1182/blood-2012-09-455436.
agulation factor VII. Thromb Haemost. 2005;94:493–497. 170. Rautou PE, Tatsumi K, Antoniak S, Owens AP III, Sparkenbaugh E,
152. Pawlinski R, Pedersen B, Erlich J, Mackman N. Role of tissue factor Holle LA, Wolberg AS, Kopec AK, Pawlinski R, Luyendyk JP, Mackman
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

in haemostasis, thrombosis, angiogenesis and inflammation: lessons N. Hepatocyte tissue factor contributes to the hypercoagulable state in
from low tissue factor mice. Thromb Haemost. 2004;92:444–450. doi: a mouse model of chronic liver injury. J Hepatol. 2016;64:53–59. doi:
10.1160/TH04-05-0309. 10.1016/j.jhep.2015.08.017.
153. Xu H, Noria F, Sandoval-Cooper MJ, Menchen H, Donahue DL, Ploplis 171. Shaver CM, Grove BS, Putz ND, Clune JK, Lawson WE, Carnahan RH,
VA, Castellino FJ. Severe deficiency of coagulation factor VII results in Mackman N, Ware LB, Bastarache JA. Regulation of alveolar procoagu-
spontaneous cardiac fibrosis in mice. J Pathol. 2009;217:362–371. doi: lant activity and permeability in direct acute lung injury by lung epi-
10.1002/path.2454. thelial tissue factor. Am J Respir Cell Mol Biol. 2015;53:719–727. doi:
154. Bode MF, Mackman N. A combined deficiency of tissue factor and PAR- 10.1165/rcmb.2014-0179OC.
4 is associated with fatal pulmonary hemorrhage in mice. Thromb Res. 172. Wang S, Reeves B, Sparkenbaugh EM, Russell J, Soltys Z, Zhang H,
2016;146:46–50. doi: 10.1016/j.thromres.2016.08.023. Faber JE, Key NS, Kirchhofer D, Granger DN, Mackman N, Pawlinski R.
155. Pedersen B, Holscher T, Sato Y, Pawlinski R, Mackman N. A balance Protective and detrimental effects of neuroectodermal cell-derived tissue
between tissue factor and tissue factor pathway inhibitor is required factor in mouse models of stroke. JCI Insight. 2016;1:e86663.
for embryonic development and hemostasis in adult mice. Blood. 173. Wun TC, Kretzmer KK, Girard TJ, Miletich JP, Broze GJ Jr. Cloning
2005;105:2777–2782. doi: 10.1182/blood-2004-09-3724. and characterization of a cDNA coding for the lipoprotein-associated
156. Erlich J, Parry GC, Fearns C, Muller M, Carmeliet P, Luther T, Mackman coagulation inhibitor shows that it consists of three tandem Kunitz-type
N. Tissue factor is required for uterine hemostasis and maintenance inhibitory domains. J Biol Chem. 1988;263:6001–6004.
of the placental labyrinth during gestation. Proc Natl Acad Sci USA. 174. Rao LV, Rapaport SI. Studies of a mechanism inhibiting the initiation of
1999;96:8138–8143. the extrinsic pathway of coagulation. Blood. 1987;69:645–651.
157. Pawlinski R, Tencati M, Holscher T, Pedersen B, Voet T, Tilley
175. Sanders NL, Bajaj SP, Zivelin A, Rapaport SI. Inhibition of tissue factor/
RE, Marynen P, Mackman N. Role of cardiac myocyte tissue fac- factor VIIa activity in plasma requires factor X and an additional plasma
tor in heart hemostasis. J Thromb Haemost. 2007;5:1693–1700. doi: component. Blood. 1985;66:204–212.
10.1111/j.1538-7836.2007.02649.x. 176. Broze GJ, Girard TJ. Tissue factor pathway inhibitor: structure-function.
158. Pawlinski R, Fernandes A, Kehrle B, et al. Tissue factor deficiency Front Biosci (Landmark Ed). 2012;17:262–280.
causes cardiac fibrosis and left ventricular dysfunction. Proc Natl Acad 177. Mast AE. Tissue factor pathway inhibitor: multiple anticoagulant activi-
Sci USA. 2002;99:15333–15338. doi: 10.1073/pnas.242501899. ties for a single protein. Arterioscler Thromb Vasc Biol. 2016;36:9–14.
159. Bastarache JA, Sebag SC, Clune JK, Grove BS, Lawson WE, Janz DR, doi: 10.1161/ATVBAHA.115.305996.
Roberts LJ II, Dworski R, Mackman N, Ware LB. Low levels of tissue 178. Huang ZF, Higuchi D, Lasky N, Broze GJ Jr. Tissue factor pathway in-
factor lead to alveolar haemorrhage, potentiating murine acute lung in- hibitor gene disruption produces intrauterine lethality in mice. Blood.
jury and oxidative stress. Thorax. 2012;67:1032–1039. doi: 10.1136/ 1997;90:944–951.
thoraxjnl-2012-201781. 179. Chan JC, Carmeliet P, Moons L, Rosen ED, Huang ZF, Broze GJ, Collen
160. Antoniak S, Tatsumi K, Hisada Y, Milner JJ, Neidich SD, Shaver CM, D, Castellino FJ. Factor VII deficiency rescues the intrauterine lethality
Pawlinski R, Beck MA, Bastarache JA, Mackman N. Tissue factor de- in mice associated with a tissue factor pathway inhibitor deficit. J Clin
ficiency increases alveolar hemorrhage and death in influenza A virus- Invest. 1999;103:475–482.
infected mice. J Thromb Haemost. 2016;14:1238–1248. doi: 10.1111/ 180. Maroney SA, Cooley BC, Ferrel JP, Bonesho CE, Nielsen LV, Johansen PB,
jth.13307. Hermit MB, Petersen LC, Mast AE. Absence of hematopoietic tissue factor
161. Getz TM, Piatt R, Petrich BG, Monroe D, Mackman N, Bergmeier W. pathway inhibitor mitigates bleeding in mice with hemophilia. Proc Natl
Novel mouse hemostasis model for real-time determination of bleeding Acad Sci USA. 2012;109:3927–3931. doi: 10.1073/pnas.1119858109.
time and hemostatic plug composition. J Thromb Haemost. 2015;13:417– 181. Chowdary P, Lethagen S, Friedrich U, Brand B, Hay C, Abdul Karim F,
425. doi: 10.1111/jth.12802. Klamroth R, Knoebl P, Laffan M, Mahlangu J, Miesbach W, Dalsgaard
162. Monroe DM, Mackman N, Hoffman M. Wound healing in hemophilia B Nielsen J, Martín-Salces M, Angchaisuksiri P. Safety and pharmacoki-
mice and low tissue factor mice. Thromb Res. 2010;125(suppl 1):S74– netics of anti-TFPI antibody (concizumab) in healthy volunteers and
S77. doi: 10.1016/j.thromres.2010.01.043. patients with hemophilia: a randomized first human dose trial. J Thromb
163. Spronk H, Wilhelm S, Heemskerk J, ten Cate H, Knetsch M, Gailani Haemost. 2015;13:743–754. doi: 10.1111/jth.12864.
D, Pawlinski R, Mackman N, Renne T. Feedback activation of factor 182. Peterson JA, Maroney SA, Mast AE. Targeting TFPI for hemophilia
XI by thrombin is essential for hemostasis in vivo. J Thromb Haemost. treatment. Thromb Res. 2016;141(suppl 2):S28–S30. doi: 10.1016/
2009;7:1–2. S0049-3848(16)30359-0.
164. Snyder LA, Rudnick KA, Tawadros R, Volk A, Tam SH, Anderson 183. Mackman N. Tissue-specific hemostasis in mice. Arterioscler Thromb Vasc
GM, Bugelski PJ, Yang J. Expression of human tissue factor under Biol. 2005;25:2273–2281. doi: 10.1161/01.ATV.0000183884.06371.52.
the control of the mouse tissue factor promoter mediates normal he- 184. Mackman N, Sawdey MS, Keeton MR, Loskutoff DJ. Murine tissue fac-
mostasis in knock-in mice. J Thromb Haemost. 2008;6:306–314. doi: tor gene expression in vivo. Tissue and cell specificity and regulation by
10.1111/j.1538-7836.2007.02833.x. lipopolysaccharide. Am J Pathol. 1993;143:76–84.
724   Arterioscler Thromb Vasc Biol   April 2018

185. Eddleston M, de la Torre JC, Oldstone MB, Loskutoff DJ, Edgington TS, 204. Toschi V, Gallo R, Lettino M, Fallon JT, Gertz SD, Fernández-Ortiz A,
Mackman N. Astrocytes are the primary source of tissue factor in the mu- Chesebro JH, Badimon L, Nemerson Y, Fuster V, Badimon JJ. Tissue
rine central nervous system. A role for astrocytes in cerebral hemostasis. factor modulates the thrombogenicity of human atherosclerotic plaques.
J Clin Invest. 1993;92:349–358. doi: 10.1172/JCI116573. Circulation. 1997;95:594–599.
186. Mullins ES, Kombrinck KW, Talmage KE, Shaw MA, Witte DP, Ullman 205. Bonderman D, Teml A, Jakowitsch J, Adlbrecht C, Gyöngyösi M, Sperker
JM, Degen SJ, Sun W, Flick MJ, Degen JL. Genetic elimination of W, Lass H, Mosgoeller W, Glogar DH, Probst P, Maurer G, Nemerson Y,
prothrombin in adult mice is not compatible with survival and results Lang IM. Coronary no-reflow is caused by shedding of active tissue fac-
in spontaneous hemorrhagic events in both heart and brain. Blood. tor from dissected atherosclerotic plaque. Blood. 2002;99:2794–2800.
2009;113:696–704. doi: 10.1182/blood-2008-07-169003. 206. Tremoli E, Camera M, Toschi V, Colli S. Tissue factor in atherosclerosis.
187. Mackman N. Bleeding hearts. Blood. 2009;113:500–501. doi: 10.1182/ Atherosclerosis. 1999;144:273–283.
blood-2008-11-188508. 207. Camera M, Toschi V, Brambilla M, Lettino M, Rossetti L, Canzano P, Di
188. Tai SJ, Herzog RW, Margaritis P, Arruda VR, Chu K, Golden JA,
Minno A, Tremoli E. The role of tissue factor in atherothrombosis and
Labosky PA, High KA. A viable mouse model of factor X deficiency coronary artery disease: insights into platelet tissue factor. Semin Thromb
provides evidence for maternal transfer of factor X. J Thromb Haemost. Hemost. 2015;41:737–746. doi: 10.1055/s-0035-1564041.
2008;6:339–345. doi: 10.1111/j.1538-7836.2007.02849.x. 208. Owens AP III, Mackman N. Role of tissue factor in atherothrombosis. Curr
189. Shaw MA, Kombrinck KW, McElhinney KE, Sweet DR, Flick MJ, Atheroscler Rep. 2012;14:394–401. doi: 10.1007/s11883-012-0269-5.
Palumbo JS, Cheng M, Esmon NL, Esmon CT, Brill A, Wagner DD, 209. Tatsumi K, Mackman N. Tissue factor and atherothrombosis. J

Degen JL, Mullins ES. Limiting prothrombin activation to meizothrom- Atheroscler Thromb. 2015;22:543–549. doi: 10.5551/jat.30940.
bin is compatible with survival but significantly alters hemostasis in 210. Owens AP III, Mackman N. Sources of tissue factor that contribute
mice. Blood. 2016;128:721–731. doi: 10.1182/blood-2015-11-680280. to thrombosis after rupture of an atherosclerotic plaque. Thromb Res.
190. Owens AP III, Mackman N. Tissue factor and thrombosis: the clot starts 2012;129(suppl 2):S30–S33. doi: 10.1016/j.thromres.2012.02.026.
here. Thromb Haemost. 2010;104:432–439. doi: 10.1160/TH09-11-0771. 211. Lechner D, Weltermann A. Circulating tissue factor-exposing mic-
191. Hatakeyama K, Asada Y, Marutsuka K, Sato Y, Kamikubo Y, Sumiyoshi roparticles. Thromb Res. 2008;122(suppl 1):S47–S54. doi: 10.1016/
A. Localization and activity of tissue factor in human aortic atheroscle- S0049-3848(08)70019-7.
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

rotic lesions. Atherosclerosis. 1997;133:213–219. 212. Mackman N. Role of tissue factor in hemostasis, thrombosis, and vas-
192. Wilcox JN, Smith KM, Schwartz SM, Gordon D. Localization of tissue cular development. Arterioscler Thromb Vasc Biol. 2004;24:1015–1022.
factor in the normal vessel wall and in the atherosclerotic plaque. Proc doi: 10.1161/01.ATV.0000130465.23430.74.
Natl Acad Sci USA. 1989;86:2839–2843. 213. Owens AP III, Mackman N. Microparticles in hemostasis and thrombosis.
193. Mallat Z, Hugel B, Ohan J, Lesèche G, Freyssinet JM, Tedgui A. Shed Circ Res. 2011;108:1284–1297. doi: 10.1161/CIRCRESAHA.110.233056.
membrane microparticles with procoagulant potential in human ath- 214. Hisada Y, Alexander W, Kasthuri R, Voorhees P, Mobarrez F, Taylor A,
erosclerotic plaques: a role for apoptosis in plaque thrombogenicity. McNamara C, Wallen H, Witkowski M, Key NS, Rauch U, Mackman N.
Circulation. 1999;99:348–353. Measurement of microparticle tissue factor activity in clinical samples: a
194. Johnson GJ, Leis LA, Bach RR. Tissue factor activity of blood mono- summary of two tissue factor-dependent FXa generation assays. Thromb
nuclear cells is increased after total knee arthroplasty. Thromb Haemost. Res. 2016;139:90–97. doi: 10.1016/j.thromres.2016.01.011.
2009;102:728–734. doi: 10.1160/TH09-04-0261. 215. Westrick RJ, Bodary PF, Xu Z, Shen YC, Broze GJ, Eitzman DT.

195. Geddings JE, Mackman N. Tumor-derived tissue factor-positive
Deficiency of tissue factor pathway inhibitor promotes atherosclerosis
microparticles and venous thrombosis in cancer patients. Blood. and thrombosis in mice. Circulation. 2001;103:3044–3046.
2013;122:1873–1880. doi: 10.1182/blood-2013-04-460139. 216. Kuijpers MJ, van der Meijden PE, Feijge MA, Mattheij NJ, May F,
196. Khorana AA, Francis CW, Menzies KE, Wang JG, Hyrien O, Hathcock Govers-Riemslag J, Meijers JC, Heemskerk JW, Renné T, Cosemans JM.
J, Mackman N, Taubman MB. Plasma tissue factor may be predictive Factor XII regulates the pathological process of thrombus formation on
of venous thromboembolism in pancreatic cancer. J Thromb Haemost. ruptured plaques. Arterioscler Thromb Vasc Biol. 2014;34:1674–1680.
2008;6:1983–1985. doi: 10.1111/j.1538-7836.2008.03156.x. doi: 10.1161/ATVBAHA.114.303315.
197. Thaler J, Ay C, Mackman N, Bertina RM, Kaider A, Marosi C, Key 217. Chi L, Gibson G, Peng YW, Bousley R, Brammer D, Rekhter M, Chen
NS, Barcel DA, Scheithauer W, Kornek G, Zielinski C, Pabinger J, Leadley R. Characterization of a tissue factor/factor VIIa-dependent
I. Microparticle-associated tissue factor activity, venous throm- model of thrombosis in hypercholesterolemic rabbits. J Thromb
boembolism and mortality in pancreatic, gastric, colorectal and Haemost. 2004;2:85–92.
brain cancer patients. J Thromb Haemost. 2012;10:1363–1370. doi: 218. Asada Y, Hara S, Tsuneyoshi A, Hatakeyama K, Kisanuki A, Marutsuka
10.1111/j.1538-7836.2012.04754.x. K, Sato Y, Kamikubo Y, Sumiyoshi A. Fibrin-rich and platelet-rich throm-
198. Cuadrado MJ, López-Pedrera C, Khamashta MA, Camps MT,
bus formation on neointima: recombinant tissue factor pathway inhibitor
Tinahones F, Torres A, Hughes GR, Velasco F. Thrombosis in pri- prevents fibrin formation and neointimal development following repeat-
mary antiphospholipid syndrome: a pivotal role for monocyte tis- ed balloon injury of rabbit aorta. Thromb Haemost. 1998;80:506–511.
sue factor expression. Arthritis Rheum. 1997;40:834–841. doi: 219. Asada Y, Marutsuka K, Hatakeyama K, Sato Y, Hara S, Kisanuki A,
10.1002/1529-0131(199705)40:5<834::AID-ART8>3.0.CO;2-#. Sumiyoshi A. The role of tissue factor in the pathogenesis of thrombosis
199. Dobado-Berrios PM, López-Pedrera C, Velasco F, Aguirre MA, Torres and atherosclerosis. J Atheroscler Thromb. 1998;4:135–139.
A, Cuadrado MJ. Increased levels of tissue factor mRNA in mononuclear 220. Gando S, Levi M, Toh CH. Disseminated intravascular coagulation. Nat
blood cells of patients with primary antiphospholipid syndrome. Thromb Rev Dis Primers. 2016;2:16037. doi: 10.1038/nrdp.2016.37.
Haemost. 1999;82:1578–1582. 221. Østerud B, Bjørklid E. The tissue factor pathway in disseminated intra-
200. Reverter JC, Tàssies D, Font J, Monteagudo J, Escolar G, Ingelmo M, vascular coagulation. Semin Thromb Hemost. 2001;27:605–617. doi:
Ordinas A. Hypercoagulable state in patients with antiphospholipid syn- 10.1055/s-2001-18866.
drome is related to high induced tissue factor expression on monocytes and 222. Taylor FB Jr, Chang A, Ruf W, Morrissey JH, Hinshaw L, Catlett R, Blick
to low free protein s. Arterioscler Thromb Vasc Biol. 1996;16:1319–1326. K, Edgington TS. Lethal E. coli septic shock is prevented by blocking tis-
201. Shet AS, Aras O, Gupta K, Hass MJ, Rausch DJ, Saba N, Koopmeiners sue factor with monoclonal antibody. Circ Shock. 1991;33:127–134.
L, Key NS, Hebbel RP. Sickle blood contains tissue factor-positive 223. Pawlinski R, Pedersen B, Schabbauer G, Tencati M, Holscher T, Boisvert
microparticles derived from endothelial cells and monocytes. Blood. W, Andrade-Gordon P, Frank RD, Mackman N. Role of tissue factor and
2003;102:2678–2683. doi: 10.1182/blood-2003-03-0693. protease-activated receptors in a mouse model of endotoxemia. Blood.
202. Geisbert TW, Young HA, Jahrling PB, Davis KJ, Kagan E, Hensley LE. 2004;103:1342–1347. doi: 10.1182/blood-2003-09-3051.
Mechanisms underlying coagulation abnormalities in ebola hemorrhagic 224. Osterud B, Due J Jr. Blood coagulation in patients with benign and ma-
fever: overexpression of tissue factor in primate monocytes/macrophages lignant tumours before and after surgery. Special reference to thrombo-
is a key event. J Infect Dis. 2003;188:1618–1629. doi: 10.1086/379724. plastin generation in monocytes. Scand J Haematol. 1984;32:258–264.
203. Funderburg NT, Mayne E, Sieg SF, Asaad R, Jiang W, Kalinowska 225. Chung J, Tabuchi N, Koyama T. The increase in tissue factor-bearing leu-
M, Luciano AA, Stevens W, Rodriguez B, Brenchley JM, Douek DC, kocytes after off-pump coronary artery bypass surgery. Thromb Haemost.
Lederman MM. Increased tissue factor expression on circulating 2008;100:952–954.
monocytes in chronic HIV infection: relationship to in vivo coagula- 226. White RH, Romano PS, Zhou H, Rodrigo J, Bargar W. Incidence and
tion and immune activation. Blood. 2010;115:161–167. doi: 10.1182/ time course of thromboembolic outcomes following total hip or knee ar-
blood-2009-03-210179. throplasty. Arch Intern Med. 1998;158:1525–1531.
Grover and Mackman   Tissue Factor in Hemostasis and Thrombosis   725

227. Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH.
245. Kornberg A, Blank M, Kaufman S, Shoenfeld Y. Induction of tissue fac-
Thromboembolism is a leading cause of death in cancer patients receiv- tor-like activity in monocytes by anti-cardiolipin antibodies. J Immunol.
ing outpatient chemotherapy. J Thromb Haemost. 2007;5:632–634. doi: 1994;153:1328–1332.
10.1111/j.1538-7836.2007.02374.x. 246. Reverter JC, Tàssies D, Font J, Khamashta MA, Ichikawa K, Cervera R,
228. Hisada Y, Mackman N. Cancer-associated pathways and biomark-
Escolar G, Hughes GR, Ingelmo M, Ordinas A. Effects of human mono-
ers of venous thrombosis. Blood. 2017;130:1499–1506. doi: 10.1182/ clonal anticardiolipin antibodies on platelet function and on tissue fac-
blood-2017-03-743211. tor expression on monocytes. Arthritis Rheum. 1998;41:1420–1427. doi:
229. Davila M, Robles-Carrillo L, Unruh D, Huo Q, Gardiner C, Sargent 10.1002/1529-0131(199808)41:8<1420::AID-ART11>3.0.CO;2-U.
IL, Adam M, Woodhams BJ, Francis JL, Bogdanov VY, Amirkhosravi 247. Perez-Sanchez C, Ruiz-Limon P, Aguirre MA, Bertolaccini ML,

A. Microparticle association and heterogeneity of tumor-derived tissue Khamashta MA, Rodriguez-Ariza A, Segui P, Collantes-Estevez E,
factor in plasma: is it important for coagulation activation? J Thromb Barbarroja N, Khraiwesh H, Gonzalez-Reyes JA, Villalba JM, Velasco F,
Haemost. 2014;12:186–196. doi: 10.1111/jth.12475. Cuadrado MJ, Lopez-Pedrera C. Mitochondrial dysfunction in antiphos-
230. Garnier D, Magnus N, Lee TH, Bentley V, Meehan B, Milsom C,
pholipid syndrome: implications in the pathogenesis of the disease and
Montermini L, Kislinger T, Rak J. Cancer cells induced to express mes- effects of coenzyme Q(10) treatment. Blood. 2012;119:5859–5870. doi:
enchymal phenotype release exosome-like extracellular vesicles carry- 10.1182/blood-2011-12-400986.
ing tissue factor. J Biol Chem. 2012;287:43565–43572. doi: 10.1074/jbc. 248. Pérez-Sánchez C, Aguirre MÁ, Ruiz-Limón P, et al. Ubiquinol effects on
M112.401760. antiphospholipid syndrome prothrombotic profile: a randomized, place-
231. Dvorak HF, Quay SC, Orenstein NS, Dvorak AM, Hahn P, Bitzer AM, bo-controlled trial. Arterioscler Thromb Vasc Biol. 2017;37:1923–1932.
Carvalho AC. Tumor shedding and coagulation. Science. 1981;212: doi: 10.1161/ATVBAHA.117.309225.
923–924. 249. Grover SP, Roubey RAS. Q10uest for new therapies to prevent antiphos-
232. Tesselaar ME, Romijn FP, Van Der Linden IK, Prins FA, Bertina RM, pholipid antibody-mediated thrombosis. Arterioscler Thromb Vasc Biol.
Osanto S. Microparticle-associated tissue factor activity: a link between 2017;37:1801–1802. doi: 10.1161/ATVBAHA.117.309955.
cancer and thrombosis? J Thromb Haemost. 2007;5:520–527. doi: 250. Mackman N, Roubey RA. Is leukocyte tissue factor the key to ve-
10.1111/j.1538-7836.2007.02369.x. nous thrombosis in antiphospholipid syndrome? J Thromb Haemost.
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

233. Manly DA, Wang J, Glover SL, Kasthuri R, Liebman HA, Key NS, 2016;14:1008–1010. doi: 10.1111/jth.13296.
Mackman N. Increased microparticle tissue factor activity in cancer pa- 251. Vega-Ostertag ME, Pierangeli SS. Mechanisms of aPL-mediated throm-
tients with venous thromboembolism. Thromb Res. 2010;125:511–512. bosis: effects of aPL on endothelium and platelets. Curr Rheumatol Rep.
doi: 10.1016/j.thromres.2009.09.019. 2007;9:190–197.
234. Manly DA, Boles J, Mackman N. Role of tissue factor in venous
252. Seshan SV, Franzke CW, Redecha P, Monestier M, Mackman N, Girardi
thrombosis. Annu Rev Physiol. 2011;73:515–525. doi: 10.1146/ G. Role of tissue factor in a mouse model of thrombotic microangiopathy
annurev-physiol-042210-121137. induced by antiphospholipid antibodies. Blood. 2009;114:1675–1683.
235. Tesselaar ME, Romijn FP, van der Linden IK, Bertina RM, Osanto S. doi: 10.1182/blood-2009-01-199117.
Microparticle-associated tissue factor activity in cancer patients with 253. Prinz N, Clemens N, Canisius A, Lackner KJ. Endosomal NADPH-
and without thrombosis. J Thromb Haemost. 2009;7:1421–1423. doi: oxidase is critical for induction of the tissue factor gene in monocytes and
10.1111/j.1538-7836.2009.03504.x. endothelial cells. Lessons from the antiphospholipid syndrome. Thromb
236. Thomas GM, Panicot-Dubois L, Lacroix R, Dignat-George F, Lombardo Haemost. 2013;109:525–531. doi: 10.1160/TH12-06-0421.
D, Dubois C. Cancer cell-derived microparticles bearing P-selectin gly- 254. Manukyan D, Müller-Calleja N, Jäckel S, Luchmann K, Mönnikes

coprotein ligand 1 accelerate thrombus formation in vivo. J Exp Med. R, Kiouptsi K, Reinhardt C, Jurk K, Walter U, Lackner KJ. Cofactor-
2009;206:1913–1927. doi: 10.1084/jem.20082297. independent human antiphospholipid antibodies induce venous thrombosis
237. Thomas GM, Brill A, Mezouar S, Crescence L, Gallant M, Dubois C, in mice. J Thromb Haemost. 2016;14:1011–1020. doi: 10.1111/jth.13263.
Wagner DD. Tissue factor expressed by circulating cancer cell-derived 255. Stravitz RT, Bowling R, Bradford RL, Key NS, Glover S, Thacker LR,
microparticles drastically increases the incidence of deep vein throm- Gabriel DA. Role of procoagulant microparticles in mediating complica-
bosis in mice. J Thromb Haemost. 2015;13:1310–1319. doi: 10.1111/ tions and outcome of acute liver injury/acute liver failure. Hepatology.
jth.13002. 2013;58:304–313. doi: 10.1002/hep.26307.
238. Geddings JE, Hisada Y, Boulaftali Y, Getz TM, Whelihan M, Fuentes R, 256. Rautou PE, Vion AC, Luyendyk JP, Mackman N. Circulating mic-

Dee R, Cooley BC, Key NS, Wolberg AS, Bergmeier W, Mackman N. roparticle tissue factor activity is increased in patients with cirrhosis.
Tissue factor-positive tumor microvesicles activate platelets and enhance Hepatology. 2014;60:1793–1795. doi: 10.1002/hep.27033.
thrombosis in mice. J Thromb Haemost. 2016;14:153–166. doi: 10.1111/ 257. Austin H, Key NS, Benson JM, Lally C, Dowling NF, Whitsett C, Hooper
jth.13181. WC. Sickle cell trait and the risk of venous thromboembolism among
239. Davila M, Amirkhosravi A, Coll E, Desai H, Robles L, Colon J, Baker blacks. Blood. 2007;110:908–912. doi: 10.1182/blood-2006-11-057604.
CH, Francis JL. Tissue factor-bearing microparticles derived from 258. Verduzco LA, Nathan DG. Sickle cell disease and stroke. Blood.

tumor cells: impact on coagulation activation. J Thromb Haemost. 2009;114:5117–5125. doi: 10.1182/blood-2009-05-220921.
2008;6:1517–1524. doi: 10.1111/j.1538-7836.2008.02987.x. 259. De Franceschi L, Cappellini MD, Olivieri O. Thrombosis and sickle
240. Hisada Y, Ay C, Auriemma AC, Cooley BC, Mackman N. Human pan- cell disease. Semin Thromb Hemost. 2011;37:226–236. doi: 10.1055/s-
creatic tumors grown in mice release tissue factor-positive microvesicles 0031-1273087.
that increase venous clot size. J Thromb Haemost. 2017;15:2208–2217. 260. Gavins FN, Russell J, Senchenkova EL, De Almeida Paula L, Damazo AS,
doi: 10.1111/jth.13809. Esmon CT, Kirchhofer D, Hebbel RP, Granger DN. Mechanisms of enhanced
241. Miyakis S, Lockshin MD, Atsumi T, Branch DW, Brey RL, Cervera R, thrombus formation in cerebral microvessels of mice expressing hemoglobin-
Derksen RH, DE Groot PG, Koike T, Meroni PL, Reber G, Shoenfeld Y, S. Blood. 2011;117:4125–4133. doi: 10.1182/blood-2010-08-301366.
Tincani A, Vlachoyiannopoulos PG, Krilis SA. International consensus 261. Chantrathammachart P, Pawlinski R. Tissue factor and thrombin in sickle
statement on an update of the classification criteria for definite antiphos- cell anemia. Thromb Res. 2012;129(suppl 2):S70–S72. doi: 10.1016/j.
pholipid syndrome (APS). J Thromb Haemost. 2006;4:295–306. doi: thromres.2012.02.038.
10.1111/j.1538-7836.2006.01753.x. 262. Sparkenbaugh EM, Chantrathammachart P, Mickelson J, van Ryn J,
242. Farmer-Boatwright MK, Roubey RA. Venous thrombosis in the antiphos- Hebbel RP, Monroe DM, Mackman N, Key NS, Pawlinski R. Differential
pholipid syndrome. Arterioscler Thromb Vasc Biol. 2009;29:321–325. contribution of FXa and thrombin to vascular inflammation in a mouse
doi: 10.1161/ATVBAHA.108.182204. model of sickle cell disease. Blood. 2014;123:1747–1756. doi: 10.1182/
243. Zhou H, Wolberg AS, Roubey RA. Characterization of monocyte tis- blood-2013-08-523936.
sue factor activity induced by IgG antiphospholipid antibodies and 263. Antoniak S, Mackman N. Multiple roles of the coagulation protease cas-
inhibition by dilazep. Blood. 2004;104:2353–2358. doi: 10.1182/blood- cade during virus infection. Blood. 2014;123:2605–2613. doi: 10.1182/
2004-01-0145. blood-2013-09-526277.
244. Sorice M, Longo A, Capozzi A, Garofalo T, Misasi R, Alessandri C, Conti 264. Geisbert TW, Hensley LE, Jahrling PB, Larsen T, Geisbert JB, Paragas
F, Buttari B, Riganò R, Ortona E, Valesini G. Anti-beta2-glycoprotein J, Young HA, Fredeking TM, Rote WE, Vlasuk GP. Treatment of Ebola
I antibodies induce monocyte release of tumor necrosis factor alpha virus infection with a recombinant inhibitor of factor VIIa/tissue factor:
and tissue factor by signal transduction pathways involving lipid rafts. a study in rhesus monkeys. Lancet. 2003;362:1953–1958. doi: 10.1016/
Arthritis Rheum. 2007;56:2687–2697. doi: 10.1002/art.22802. S0140-6736(03)15012-X.
Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis
Downloaded from http://atvb.ahajournals.org/ by guest on April 25, 2018

Steven P. Grover and Nigel Mackman

Arterioscler Thromb Vasc Biol. 2018;38:709-725; originally published online February 8, 2018;
doi: 10.1161/ATVBAHA.117.309846
Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272
Greenville Avenue, Dallas, TX 75231
Copyright © 2018 American Heart Association, Inc. All rights reserved.
Print ISSN: 1079-5642. Online ISSN: 1524-4636

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://atvb.ahajournals.org/content/38/4/709

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the
Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for
which permission is being requested is located, click Request Permissions in the middle column of the Web
page under Services. Further information about this process is available in the Permissions and Rights
Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online
at:
http://atvb.ahajournals.org//subscriptions/

View publication stats

Das könnte Ihnen auch gefallen