Combinations of proinflammatory and procoagulant reactions are the unifying principle for a variety of disorders affecting the cardiovascular system. The factor XII–driven contact system starts coagulation and inflammatory mechanisms via the intrinsic pathway of coagulation and the bradykinin-producing kallikrein-kinin system, respectively. The biochemistry of the contact system in vitro is well understood; however, its in vivo functions are just beginning to emerge. Challenging the concept of the coagulation balance, targeting factor XII or its activator polyphosphate, provides protection from thromboembolic diseases without interfering with hemostasis. This suggests that the polyphosphate/factor XII axis contributes to thrombus formation while being dispensable for hemostatic processes. In contrast to deficiency in factor XII providing safe thromboprotection, excessive FXII activity is associated with the life-threatening inflammatory disorder hereditary angioedema. The current review summarizes recent findings of the polyphosphate/factor XII–driven contact system at the intersection of procoagulant and proinflammatory disease states. Elucidating the contact system offers the exciting opportunity to develop strategies for safe interference with both thrombotic and inflammatory disorders.

The crosstalk between coagulation and inflammation constitutes the unifying principle for a variety of disorders affecting the cardiovascular system. Elucidating the role of inflammation in coagulation and vice versa will introduce new perspectives to improve diagnostics and therapies for both disease entities, leading to improved patient care. The factor XII (FXII)–driven contact system and its activator polyphosphate (polyP) can serve as a paradigm that connects coagulation and inflammation. Here, we review novel roles of the polyP/FXII axis in thromboinflammatory diseases and its therapeutic implications when targeting its functions.

The plasma contact system is a procoagulant and proinflammatory protease cascade that is initiated by FXII, in a reaction involving high-molecular-weight kininogen (HK) and plasma kallikrein (PK) (Figure 1). FXII is the plasma zymogen form of the serine protease factor XIIa (FXIIa). FXII is activated to FXIIa following binding (“contact”) to negatively charged artificial or biologic surfaces (contact activation). Alternatively, active PK has the capacity to convert FXII zymogen to the active protease. During both contact-triggered autoactivation and PK-mediated heteroactivation, FXII zymogen undergoes limited proteolysis. FXII was first recognized as essential for surface-activated diagnostic blood coagulation assays (eg, the activated partial thromboplastin time) that are commonly used as a clinical measure of global plasma coagulation.1-3  FXII also has limited proteolytic activity, when bound to contact-activating surfaces without undergoing proteolysis. This suggests that surface binding is sufficient for inducing a conformation with some enzymatic activity that is amplified by proteolytic cleavage of the FXII zymogen form.4,5  The plasma contact system components FXII and HK assemble on cell surface proteoglycans of various cardiovascular cells, with the latter 1 bound to FXI or PK.6-9  In addition, PK also binds to proteoglycans in a HK-independent manner.10  Locally produced FXIIa initiates the intrinsic pathway of coagulation via its substrate factor XI (FXI) and leads to liberation of the proinflammatory mediator bradykinin (BK) by PK-mediated cleavage of HK.11  Serpin C1 esterase inhibitor (C1INH) is the major plasma inhibitor of FXIIa and PK. Deficiency or a dysfunctional C1INH is associated with a BK-mediated life-threatening inherited swelling disorder, hereditary angioedema (HAE) type I or II, respectively.12-14 

Figure 1.

Thromboinflammatory activities of FXII in vivo. Contact system proteins FXII, HK, and PK assemble on cell surfaces. Contact with surface-exposed nonsoluble and plasma-borne soluble contact activators, including polyP or misfolded protein aggregates (PAgg) and heparin (Hep) or oversulfated chondroitin sulfate (OSCS), respectively, leads to the active protease FXIIa. FXIIa in turn triggers thrombosis via the FXI-mediated intrinsic coagulation pathway. FXIIa also activates PK that produces BK from HK. BK activates kinin B2 receptors (B2R), whereas the BK metabolite des-Arg9 BK binds to kinin B1 receptors (B1R), both of which drive inflammation. C1INH regulates FXIIa and PK enzymatic activity.

Figure 1.

Thromboinflammatory activities of FXII in vivo. Contact system proteins FXII, HK, and PK assemble on cell surfaces. Contact with surface-exposed nonsoluble and plasma-borne soluble contact activators, including polyP or misfolded protein aggregates (PAgg) and heparin (Hep) or oversulfated chondroitin sulfate (OSCS), respectively, leads to the active protease FXIIa. FXIIa in turn triggers thrombosis via the FXI-mediated intrinsic coagulation pathway. FXIIa also activates PK that produces BK from HK. BK activates kinin B2 receptors (B2R), whereas the BK metabolite des-Arg9 BK binds to kinin B1 receptors (B1R), both of which drive inflammation. C1INH regulates FXIIa and PK enzymatic activity.

Close modal

Despite its importance for fibrin formation in vitro, for decades FXII had been considered to have no function for physiological hemostasis in vivo. This premise was based on the fact that FXII-deficient patients have a normal hemostatic capacity and do not suffer from spontaneous or injury-related increased bleeding. Thus, the contact system was dormant for 50 years but recently experienced a revival.

Our laboratory has generated the first FXII-deficient (FXII−/−) mouse model and reported in 2005 that occlusive thrombus formation in both arterial and venous beds was severely defective in these animals. However, similar to FXII-deficient individuals, the hemostatic potential of FXII−/− mice remained unaffected.15  Reconstitutions of the animals with human FXII restored the defective thrombus formation, indicating a critical function of human FXII in pathological clotting. FXII−/− mice and wild-type mice treated with FXIIa inhibitors were also protected from thromboembolic diseases, including stroke, atherothrombosis, and pulmonary embolism.16-18  The importance of FXII in thrombosis is not restricted to mice. Independent groups have confirmed the critical role that the FXII-driven intrinsic pathway of coagulation plays in thrombosis in large animals such as rats,19  rabbits,20,21  or baboons.22  Furthermore, targeting the FXIIa substrate FXI in the intrinsic pathway of coagulation has proven to be a safe therapeutic strategy in recent human studies on venous thrombosis.23  Previous work also had shown that mice deficient in the FXIIa substrate of the intrinsic pathway of coagulation FXI were protected from arterial thrombosis in an identical manner.24  Initially, defective FXI activation by tissue factor–produced thrombin (“feedback activation loop”)25,26  was proposed as the mechanistic basis underlying impaired thrombus formation in FXI−/− mice. However, the observation that occlusive thrombus formation was similarly defective in FXII−/−, FXI−/− mice, as well as in animals with combined deficiency in both clotting factors (FXII−/−/FXI−/−), indicates that FXI is mostly activated by FXIIa in pathological platelet-mediated thrombosis.18  Supporting a role of the FXII-driven contact system in thrombosis, mice with deficiencies in PK27,28  or HK29  are also protected from thrombosis. A more comprehensive overview about targeting FXII and contact system proteins in various experimental thrombosis models in animals is presented in a recent review.30 

The selective importance of FXII in pathologic thrombosis raises the exciting possibility that targeting FXII is an effective strategy for the prevention and treatment of pathologic thrombosis without the bleeding risk that is characteristic of current anticoagulants. The recombinant FXIIa inhibitor rHA-infestin-4 of insect origin largely reduced arterial and venous thrombosis and offered protection from pulmonary embolism, atherothrombosis, and ischemic stroke in rodent models without a therapy-associated increase in bleeding. Furthermore, rHA-infestin-4–treated mice were protected from experimental autoimmune encephalomyelitis31,32  and hypotonic shock in anaphylaxis.33  However, rHA-infestin-4 also inhibits plasmin and FXa at high concentrations and has immunogenic properties that limit its potential clinical applications.19,32  A fully human FXIIa neutralizing antibody, 3F7, provided protection from thrombosis in a medically meaningful setting (an experimental model of extracorporeal membrane oxygenation system in rabbits) as effective as heparin. Most importantly, unlike heparin, 3F7 did not increase bleeding.21  Consistent with the importance of the FXII-FXI axis in thrombosis, targeting activated FXI (FXIa), using an antibody against the FXIa enzymatic pocket (named DEF), interfered with thread-induced venous thrombosis in a rabbit model. In contrast to FXII, deficiency in FXI is associated with a bleeding disorder (hemophilia C). For potential interference with neutralizing FXIa-associated bleedings, a reversal agent (a human immunoglobulin G) was developed that rapidly reversed DEF antibody activity in human plasma and in vivo in rabbits.34  A summary on targeting FXI for thromboprotection is presented in an excellent review article.35  An inhibitor-independent alternative approach for FXII inhibition is based on antisense oligonucleotides–mediated interference with expression of the coagulation factor. Repetitive antisense oligonucleotides injections reduced arterial and venous thrombosis in mice to levels of <25% of normal36  and attenuated catheter-induced thrombosis in rabbits20  without increased therapy-associated bleeding. An overview on currently available experimental FXII inhibitors can be found in recent reviews.37-39 

In contrast to the definite findings in animal models, impact of contact system deficiency states for human disease has remained less clear mostly due to small “patient” numbers and lack of matched controls.40  Human deficiency in FXII, PK, or HK is not associated with bleeding symptoms, although FXI deficiency is associated with mild bleeding, mainly in tissues with high fibrinolytic activity. Based on >500 individuals, severe FXI deficiency has been associated with lower risk for cardiovascular and venous thromboembolism events within the Ashkenazi Jewish community.41  Although FXI deficiency impairs hemostasis in this specific population, the data support a role for the intrinsic pathway protease in thrombosis in humans. To associate FXII deficiency with disease states, we are establishing a “patient” registry, including inherited severe FXII deficiencies (<10%; www.factor12.net) and invite readers to register their FXII-deficient “patients” there. The registry may help in assessing prevalence, incidence, severity, and characteristics of inflammatory, infectious, and thromboembolic diseases in the absence of contact activation. Together, these studies established an essential role of the FXII-driven intrinsic pathway of coagulation in thrombosis and led to a paradigm shift in the design of anticoagulant therapies pursuing the strategy of FXII inhibition as a safe therapeutic approach for the treatment of thrombotic diseases.

For more than half a century, it has been known that activated platelets promote coagulation in an FXII-dependent manner,42  and multiple groups have confirmed that platelets initiate coagulation in an FXII-dependent manner.43-45  The platelet-derived FXII activator, however, had remained obscure. We have identified polyP (Figure 1) as a platelet-derived FXII contact activator in vivo.18  PolyP initiates FXII contact activation in human plasma,46  with critical implications for thrombosis in vivo.18  The data extended on earlier findings, which had analyzed procoagulant activities of polyP in vitro, and found that platelet-size polyP activates FXII-dependent coagulation ex vivo.46  Oppositely, FXII-dependent plasma clotting and thrombus formation are defective in mice and humans with genetic deficiency in platelet polyP (inositol hexakisphosphate kinase 1 null [Ip6k1−/−] mice47  and Hermansky-Pudlak syndrome,18  respectively). Similar to whole platelets, membrane vesicles shed from activated platelets (microparticles) stimulate coagulation in an FXII-dependent manner.48  In addition to inducing FXII contact activation, polyP participates in an array of other procoagulant reactions involving fibrin, tissue factor pathway inhibitor, von Willebrand factor, and factors V and XI. Although polyP has the capacity to modulate these various pathways ex vivo, the in vivo relevance of the polymer in these mechanisms has remained unknown.49,50  PolyP procoagulant activity increases with chain length. Plasma-soluble polymers have a mean chain length of 60 to 100 and have only limited capacity to trigger FXII contact activation51 ; however, they strongly interfere with tissue factor pathway inhibitor activities in buffer systems.52  The fact that activated platelets potently activate FXII suggests that distinct polyP pools exist in platelets (Figure 2). Platelets store polyP together with high concentrations of calcium ions (>2 molar) in intracellular organelles (dense granules), and released polyP is complexed with Ca2+ ions.53  When Ca2+-polyP nanoparticles are formed under physiological conditions, these are stable for hours.54  A small fraction of platelet polyP only is secreted into the supernatant upon activation; however, the vast majority of platelet polyP is retained on procoagulant platelet cell surface upon secretion in the form of Ca2+-rich nanoparticles.55  These polyP nanoparticles potently initiate FXII contact activation and drive coagulation via the intrinsic pathway on the platelet surface.55  Similar to kaolin (a silicate that is commonly used to trigger FXII activation in the activated partial thromboplastin time clotting assay), Ca2+-polyP nanoparticles are not soluble and accumulate on the platelet membranes via unidentified cell surface binding sites.56  Ca2+-polyP nanoparticles appear to represent the natural kaolin that initiates the contact system on cardiovascular surfaces. Similar to polyP nanoparticles, short-chain polyP that is conjugated to colloidal gold nanoparticles activates FXII to a similar extent as compared with long-chain polyP.57  Consistently, localization onto particles increases procoagulant activity of short-chain polyP even under flow conditions.58  The procoagulant properties of insoluble polyP packed in nanoparticles largely differ from those of molecularly dissolved soluble molecules. FXII-activating properties of polymers in nanoparticle form largely exceed that of dispersed polyP in solution.54,56  The formation of Ca2+-polyP aggregates with increased capacity for inducing FXII contact activation argues against a decisive role of polymer chain length in regulating polyP activity in vivo. Synthetic long-chain polyP binds to the platelet surface, whereas short-chain polyP does not.59  This could indicate that polyP microparticles on the platelet surface contain longer polymers than are found in the supernatant of activated platelets or condensed aggregated short-chain molecules (Figure 2). Taken together, the polyP/FXII-driven coagulation has a critical function for thrombosis albeit does not contribute to hemostatic processes (Figure 3). The findings with polyP/FXII also suggest that the classical concept of a coagulation balance with hemostasis and thrombosis presenting the opposite poles has some exceptions.

Figure 2.

Activities and functions of polyP in the coagulation system. Two different pools of polyP that differ in their biophysical properties and procoagulant mechanisms, respectively, operate in the coagulation cascade. Insoluble long-chain polyP and polymers condensed in microparticles initiate FXII contact activation on cell surfaces. Although mechanisms driven by insoluble polyP microparticles have been analyzed in vivo, functions of soluble/short-chain polymers are based on in vitro studies.

Figure 2.

Activities and functions of polyP in the coagulation system. Two different pools of polyP that differ in their biophysical properties and procoagulant mechanisms, respectively, operate in the coagulation cascade. Insoluble long-chain polyP and polymers condensed in microparticles initiate FXII contact activation on cell surfaces. Although mechanisms driven by insoluble polyP microparticles have been analyzed in vivo, functions of soluble/short-chain polymers are based on in vitro studies.

Close modal
Figure 3.

The polyP/FXII axis in thrombosis. At a site of vessel injury, tissue factor–initiated coagulation mechanisms mediate hemostasis, and deficiency in these pathways results in bleeding. In thrombosis, activated platelets expose polyP nanoparticles that induce FXII contact activation driving coagulation within the thrombus. The polyP/FXII specifically contributes to pathologic coagulation, and targeting polyP or FXII interferes with thrombosis but has no impact on hemostasis, suggesting use of this strategy for safe thromboprotection.

Figure 3.

The polyP/FXII axis in thrombosis. At a site of vessel injury, tissue factor–initiated coagulation mechanisms mediate hemostasis, and deficiency in these pathways results in bleeding. In thrombosis, activated platelets expose polyP nanoparticles that induce FXII contact activation driving coagulation within the thrombus. The polyP/FXII specifically contributes to pathologic coagulation, and targeting polyP or FXII interferes with thrombosis but has no impact on hemostasis, suggesting use of this strategy for safe thromboprotection.

Close modal

Based on the potent procoagulant activities of polyP, the polymer has emerged as a novel target for anticoagulant drugs. Several strategies to achieve this aim have been developed. First, recombinant salivary proteins of the African sand fly (PdSP15) bind polyP and interfere with FXII contact system–driven clotting and inflammation. However, PdSP15 binds to various other polyanions, such as silica, limiting its potential therapeutic application.60  Second, cationic polyethylenimine and polyamidoamine dendrimers bind polyP and attenuate thrombosis in mouse models.61,62  Unfortunately, polyethylenimine and polyamidoamine at a concentration required to provide thromboprotection is cytotoxic to cultured cells. Third, the crown ether-based universal heparin reversal agents (UHRAs that originally were developed as heparin antidotes) are nontoxic and inhibit polyP procoagulant activity. However, UHRAs also interfere with tissue factor–initiated fibrin formation,63  and UHRA infusions lead to bleeding in murine models,63  involving charge-driven interference with factor V activation. Finally, recombinant Escherichia coli exopolyphosphatase (PPX, an enzyme that specifically degrades polyP of chain length >35 phosphate units) mutants even at the highest concentrations tested selectively targeted polyP. PolyP binding by truncated mutant PPX_Δ12 or degradation by full-size PPX, respectively, almost completely blunted fibrin formation on activated platelets and ablated arterial and venous thrombosis in mouse models in an FXIIa-dependent manner. Supporting the notion that polyP functions via FXII contact activation in vivo, specific neutralization of polyP interfered with thrombosis but did not impair the hemostatic capacity of treated animals, reproducing the thromboprotective phenotype of FXII-deficient animals. Targeting polyP in the circulation and at platelet surfaces for interference with thrombosis and inflammation in vivo is a matter of ongoing research. The nucleotide RNA is negatively charged and has the capacity for inducing FXII contact activation ex vivo.64  Extracellular RNA has been considered to promote thrombosis in vivo in an FXII-dependent manner. This notion is based on the observation that infusion of RNase (an enzyme that degrades RNA) interferes with arterial thrombosis in a murine arterial thrombosis model.65  However, RNase also readily hydrolyzes the phosphoanhydride bonds in the polyP backbone and efficiently degrades the polymer, offering an alternative explanation for the thromboprotective effects conferred by the enzyme.55  Targeting both polyP and FXII reduces mechanical stability of the clot structure within the thrombus in vitro by interference with fibrin fiber formation.18,46  Oppositely, interference with FXIIa-driven coagulation increases embolization events following atherothrombosis, confirming a role of polyP/FXII for thrombus propagation distant from the vessel wall but not in coagulation mechanisms at the site of vascular injury.17,66 

The importance of polyP in thrombosis is not restricted to platelets. Cancer cells and cancer-derived microparticles expose polyP on their surface that initiates pathologic clotting in an FXII-dependent manner in murine venous thrombosis models and patients with malignancies,67  suggesting that targeting polyP could be a novel therapy in a variety of disease states associated with increased thrombotic risk. PolyP is not stable, and the polymer is degraded with a 2-hour half-life in plasma.46  Furthermore, purified polyP loses its procoagulant activity during storage.68  The polyP/FXII pathway operates independently of tissue factor–mediated coagulation and triggers thrombin formation even when the tissue factor pathway is inhibited.68  Oppositely, FXII−/− mice are protected from tissue factor–infusion triggered thrombosis. Independent of coagulation, polyP potently triggers inflammatory activities in rodent models. FXIIa that is generated following binding to polyP activates the kallikrein-kinin system, resulting in BK generation in vitro and BK-driven edema in vivo.30  Furthermore, polyP modulates the complement system69  and can activate the mammalian target of rapamycin in breast cancer cells and thus stimulate mitogenic signaling pathways.70  The polymer also forms high-affinity complexes with high mobility group box 1 (HMGB1) and histone 471 ; both proteins are increased in serum/plasma of septic patients.72  PolyP binding to these proteins amplifies proinflammatory signaling in endothelial cells through interaction and activation of 2 cell surface receptors, RAGE and P2Y1,71  leading to disruption of endothelial barriers by mammalian target of rapamycin–dependent pathways.73  Furthermore, platelet polyP binding to histones initiates thrombin formation independent of contact activation.74  To date, little is known about polyP biosynthesis and regulation in mammalian systems; however, the recently established fluorescence-activated cell sorting assay to measure polyP in patient samples based on a recombinant polyP-specific probe (PPX_Δ12) may open the avenue to establish the polymer as a novel biomarker.61 

Because of its original identification as part of the coagulation mechanism, the inflammatory branch of the contact system has long been underappreciated. However, quantitative and qualitative C1INH deficiency (C1INH-HAE; also termed type I and II HAE) results in aggressive attacks of tissue swelling in the rare disease HAE. These swelling attacks are caused by excessive production of BK as a result of a hyperactive contact activation system.11  BK acts on kinin receptors that are constitutively present on the endothelium (kinin B2 receptors) and can be expressed by leukocytes during inflammatory reactions (kinin B1 receptors). Kinin receptor signaling (among others) increases vascular leakage. BK has a half-life of seconds because of the rapid breakdown by kininases (enzymes that break down kinins) in blood and tissue. Angiotensin-converting enzyme is an important kininase and angiotensin-converting enzyme inhibitor use is known to cause angioedema (without C1INH deficiency) as a rare side effect. Murine models of C1INH deficiency have recapitulated the phenotype of excessive BK-mediated vascular leakage that characterizes HAE.75,76  Attacks of angioedema can occur throughout the body and are often seen in mucosal tissues, including the face and gastrointestinal tract as well as genitals and extremities. When the upper airways are threatened, it can have life-threatening consequences. Although attacks can travel from 1 site in the body to another (sometimes 2 sites are simultaneously affected),77  there is a remarkable restriction to the extent of vascular leakage. In other words, there is little evidence for a systemically increased vascular permeability as blood pressure remains largely normal. There are 2 complementary explanations for the restricted localization of tissue swelling: (1) The affected tissues are “primed” by locals threats (injury or infection) leading to kinin B1 receptor expression. Systemic BK production (which should happen constitutively) becomes locally visible as a result.78  (2) Analogous to blood coagulation, factors of the contact system assemble on the vascular endothelium,7  or in the vicinity of activated leukocytes and mast cells. BK is locally produced and acts on nearby tissue in a paracrine manner.8  Kininases prevent systemic action of this potent proinflammatory peptide. A particularly interesting feature of HAE is that there is a striking lack of prothrombotic features, suggesting that BK production can be initiated in the absence of (clinically significant) coagulation.79  The mechanisms that drive this mechanistic uncoupling of FXIIa-driven thrombosis and inflammation deserve further scientific attention. All FXII contact activators identified so far either trigger both coagulation and inflammation or inflammation alone but not specifically coagulation without inflammation.80  BK-driven tissue swelling attacks can also take place when C1INH levels are normal and with equally dangerous consequences. In a subset of HAE patients, the disease is not caused by C1INH deficiency, but by gain-of-function mutations in the F12 gene (encodes FXII) instead (FXII-HAE/type III HAE81 ). The clinical phenotype of these patients during attacks is very comparable with those that have C1INH-dependent HAE, although the estrogen dependence of the disease is more prominent.82  Furthermore, excessive BK production is observed in patients with angioedema of unknown origin,83  and BK-driven edema accompanies severe allergic reactions.33  These observations indicate that imbalances in the plasma contact system have pathological consequences. The associated spontaneous pathology has inflammatory features, rather than thrombotic features.

We recently performed detailed investigation of the FXII-HAE mutations, all of which are located in the proline-rich region that is unique to FXII. We first discovered that the mutations cause the production of a functional FXII protein that has a glycosylation defect.79  This reduces the net charge of the protein, promoting association to the contact system-activating polymer dextran sulfate. In vivo expression of FXII-HAE mutants in mice results in an increased propensity to develop edema in a provocation model. We subsequently identified that these mutations also render the FXII molecule sensitive to activating cleavage by plasmin.84  Although not considered part of the contact system in vitro, plasmin has a known capacity for FXII activation.85  In addition, recent studies indicate that soluble polyP enhances the plasminogen activator function of FXIIa.86  Furthermore, BK signaling contributes to blood-brain barrier permeability in hyperfibrinolytic states,87  and oppositely, antifibrinolytic agents have therapeutic value in HAE and idiopathic angioedema.88  Together, this implicates plasmin as an active player in activation of the contact system during BK production in HAE. More recent studies indicate that the interaction between the fibrinolytic system and the contact system extends beyond HAE. Brain edema is a clinically relevant side effect of thrombolytic therapy, but is considered a necessary evil to clear obstructive thrombi. It was recently demonstrated that BK is an essential player in the development of brain edema that follows plasmin activity.87  Other studies demonstrated the involvement of PK in the development of thrombolysis-associated brain edema89  and confirmed the exciting possibility that targeting the contact system eliminates this dangerous side effect of this essential therapy.

In this review, we provided an overview on the contact system and its activators in thrombosis and BK-mediated inflammatory reactions. Since the original discovery that FXII is essential for “pathological” thrombosis in 2005, the contact system and its activator polyP have been recognized to critically contribute to thromboembolic and inflammatory disorders. Targeting FXIIa and polyP interferes with the polyP/FXII axis, providing safe thromboprotection with additional anti-inflammatory activities. Because polyP/FXII inhibition potently protects from thrombosis without increasing bleeding risk, the proposed studies have the potential to improve the benefit-to-risk profile of anticoagulant therapy in comparison with currently available agents (heparins, vitamin K antagonists, factor Xa inhibitors, or thrombin inhibitors). We propose that therapeutic targeting of FXII/FXIIa has an optimal safety profile for contact system–driven thrombosis. Furthermore, therapeutic interference with the contact system factors has the additional benefit of having anti-inflammatory90  and healing91  potential. Targeting these combined features appears as the ultimate strategy to interfere with thromboinflammation. Furthermore, these studies will lay the foundation for a more complete understanding of how targeting polyP and FXII may serve as a key strategy to improving infectious, autoimmune, and allergic diseases.

The authors thank all the researchers that they had the pleasure to collaborate with in the last 12 years of exciting research with FXII.

This work was supported in part by grants from the German Research Society (SFB877, TP A11, SFB841, TP B8), and a European Research Council grant (ERC-StG-2012-311575_F-12) (T.R.). C.M. gratefully acknowledges financial support from the Landsteiner Foundation for Blood Transfusion Research and the Netherlands Thrombosis Foundation.

Contribution: T.R. and C.M. wrote the review and discussed its contents.

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

Correspondence: Thomas Renné, Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany; e-mail: thomas@renne.net.

1.
Long
AT
,
Kenne
E
,
Jung
R
,
Fuchs
TA
,
Renné
T
.
Contact system revisited: an interface between inflammation, coagulation, and innate immunity
.
J Thromb Haemost
.
2016
;
14
(
3
):
427
-
437
.
2.
Nickel
KF
,
Long
AT
,
Fuchs
TA
,
Butler
LM
,
Renné
T
.
Factor XII as a therapeutic target in thromboembolic and inflammatory diseases
.
Arterioscler Thromb Vasc Biol
.
2017
;
37
(
1
):
13
-
20
.
3.
Weitz
JI
,
Fredenburgh
JC
.
Factors XI and XII as targets for new anticoagulants
.
Front Med (Lausanne)
.
2017
;
4
:
19
.
4.
Engel
R
,
Brain
CM
,
Paget
J
,
Lionikiene
AS
,
Mutch
NJ
.
Single-chain factor XII exhibits activity when complexed to polyphosphate
.
J Thromb Haemost
.
2014
;
12
(
9
):
1513
-
1522
.
5.
Ivanov
I
,
Matafonov
A
,
Sun
MF
, et al
.
Proteolytic properties of single-chain factor XII: a mechanism for triggering contact activation
.
Blood
.
2017
;
129
(
11
):
1527
-
1537
.
6.
Renné
T
,
Müller-Esterl
W
.
Cell surface-associated chondroitin sulfate proteoglycans bind contact phase factor H-kininogen
.
FEBS Lett
.
2001
;
500
(
1-2
):
36
-
40
.
7.
Renné
T
,
Dedio
J
,
David
G
,
Müller-Esterl
W
.
High molecular weight kininogen utilizes heparan sulfate proteoglycans for accumulation on endothelial cells
.
J Biol Chem
.
2000
;
275
(
43
):
33688
-
33696
.
8.
Renné
T
,
Schuh
K
,
Müller-Esterl
W
.
Local bradykinin formation is controlled by glycosaminoglycans
.
J Immunol
.
2005
;
175
(
5
):
3377
-
3385
.
9.
Wujak
L
,
Didiasova
M
,
Zakrzewicz
D
,
Frey
H
,
Schaefer
L
,
Wygrecka
M
.
Heparan sulfate proteoglycans mediate factor XIIa binding to the cell surface
.
J Biol Chem
.
2015
;
290
(
11
):
7027
-
7039
.
10.
Veronez
CL
,
Nascimento
FD
,
Melo
KR
,
Nader
HB
,
Tersariol
IL
,
Motta
G
.
The involvement of proteoglycans in the human plasma prekallikrein interaction with the cell surface
.
PLoS One
.
2014
;
9
(
3
):
e91280
.
11.
Hofman
Z
,
de Maat
S
,
Hack
CE
,
Maas
C
.
Bradykinin: inflammatory product of the coagulation system
.
Clin Rev Allergy Immunol
.
2016
;
51
(
2
):
152
-
161
.
12.
Renné
T
.
The procoagulant and proinflammatory plasma contact system
.
Semin Immunopathol
.
2012
;
34
(
1
):
31
-
41
.
13.
Björkqvist
J
,
Sala-Cunill
A
,
Renné
T
.
Hereditary angioedema: a bradykinin-mediated swelling disorder
.
Thromb Haemost
.
2013
;
109
(
3
):
368
-
374
.
14.
Wu
MA
,
Perego
F
,
Zanichelli
A
,
Cicardi
M
.
Angioedema phenotypes: disease expression and classification
.
Clin Rev Allergy Immunol
.
2016
;
51
(
2
):
162
-
169
.
15.
Renné
T
,
Pozgajová
M
,
Grüner
S
, et al
.
Defective thrombus formation in mice lacking coagulation factor XII
.
J Exp Med
.
2005
;
202
(
2
):
271
-
281
.
16.
Kleinschnitz
C
,
Stoll
G
,
Bendszus
M
, et al
.
Targeting coagulation factor XII provides protection from pathological thrombosis in cerebral ischemia without interfering with hemostasis
.
J Exp Med
.
2006
;
203
(
3
):
513
-
518
.
17.
Kuijpers
MJ
,
van der Meijden
PE
,
Feijge
MA
, et al
.
Factor XII regulates the pathological process of thrombus formation on ruptured plaques
.
Arterioscler Thromb Vasc Biol
.
2014
;
34
(
8
):
1674
-
1680
.
18.
Müller
F
,
Mutch
NJ
,
Schenk
WA
, et al
.
Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo
.
Cell
.
2009
;
139
(
6
):
1143
-
1156
.
19.
Xu
Y
,
Cai
TQ
,
Castriota
G
, et al
.
Factor XIIa inhibition by Infestin-4: in vitro mode of action and in vivo antithrombotic benefit
.
Thromb Haemost
.
2014
;
111
(
4
):
694
-
704
.
20.
Yau
JW
,
Liao
P
,
Fredenburgh
JC
, et al
.
Selective depletion of factor XI or factor XII with antisense oligonucleotides attenuates catheter thrombosis in rabbits
.
Blood
.
2014
;
123
(
13
):
2102
-
2107
.
21.
Larsson
M
,
Rayzman
V
,
Nolte
MW
, et al
.
A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk
.
Sci Transl Med
.
2014
;
6
(
222
):
222ra17
.
22.
Matafonov
A
,
Leung
PY
,
Gailani
AE
, et al
.
Factor XII inhibition reduces thrombus formation in a primate thrombosis model
.
Blood
.
2014
;
123
(
11
):
1739
-
1746
.
23.
Büller
HR
,
Bethune
C
,
Bhanot
S
, et al
;
FXI-ASO TKA Investigators
.
Factor XI antisense oligonucleotide for prevention of venous thrombosis
.
N Engl J Med
.
2015
;
372
(
3
):
232
-
240
.
24.
Rosen
ED
,
Gailani
D
,
Castellino
FJ
.
FXI is essential for thrombus formation following FeCl3-induced injury of the carotid artery in the mouse
.
Thromb Haemost
.
2002
;
87
(
4
):
774
-
776
.
25.
Naito
K
,
Fujikawa
K
.
Activation of human blood coagulation factor XI independent of factor XII. Factor XI is activated by thrombin and factor XIa in the presence of negatively charged surfaces
.
J Biol Chem
.
1991
;
266
(
12
):
7353
-
7358
.
26.
Gailani
D
,
Broze
GJ
Jr
.
Factor XI activation in a revised model of blood coagulation
.
Science
.
1991
;
253
(
5022
):
909
-
912
.
27.
Bird
JE
,
Smith
PL
,
Wang
X
, et al
.
Effects of plasma kallikrein deficiency on haemostasis and thrombosis in mice: murine ortholog of the Fletcher trait
.
Thromb Haemost
.
2012
;
107
(
6
):
1141
-
1150
.
28.
Göb
E
,
Reymann
S
,
Langhauser
F
, et al
.
Blocking of plasma kallikrein ameliorates stroke by reducing thromboinflammation
.
Ann Neurol
.
2015
;
77
(
5
):
784
-
803
.
29.
Merkulov
S
,
Zhang
WM
,
Komar
AA
, et al
.
Deletion of murine kininogen gene 1 (mKng1) causes loss of plasma kininogen and delays thrombosis
.
Blood
.
2008
;
111
(
3
):
1274
-
1281
.
30.
Kenne
E
,
Nickel
KF
,
Long
AT
, et al
.
Factor XII: a novel target for safe prevention of thrombosis and inflammation
.
J Intern Med
.
2015
;
278
(
6
):
571
-
585
.
31.
Göbel
K
,
Pankratz
S
,
Asaridou
CM
, et al
.
Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells
.
Nat Commun
.
2016
;
7
:
11626
.
32.
May
F
,
Krupka
J
,
Fries
M
, et al
.
FXIIa inhibitor rHA-Infestin-4: safe thromboprotection in experimental venous, arterial and foreign surface-induced thrombosis
.
Br J Haematol
.
2016
;
173
(
5
):
769
-
778
.
33.
Sala-Cunill
A
,
Bjorkqvist
J
,
Senter
R
, et al
.
Plasma contact system activation drives anaphylaxis in severe mast cell-mediated allergic reactions
.
J Allergy Clin Immunol
.
2015
;
135
(
4
):
1031
-
1043.e6
.
34.
David
T
,
Kim
YC
,
Ely
LK
, et al
.
Factor XIa-specific IgG and a reversal agent to probe factor XI function in thrombosis and hemostasis
.
Sci Transl Med
.
2016
;
8
(
353
):
353ra112
.
35.
Gailani
D
,
Gruber
A
.
Factor XI as a therapeutic target
.
Arterioscler Thromb Vasc Biol
.
2016
;
36
(
7
):
1316
-
1322
.
36.
Revenko
AS
,
Gao
D
,
Crosby
JR
, et al
.
Selective depletion of plasma prekallikrein or coagulation factor XII inhibits thrombosis in mice without increased risk of bleeding
.
Blood
.
2011
;
118
(
19
):
5302
-
5311
.
37.
Kenne
E
,
Renné
T
.
Factor XII: a drug target for safe interference with thrombosis and inflammation
.
Drug Discov Today
.
2014
;
19
(
9
):
1459
-
1464
.
38.
Fredenburgh
JC
,
Gross
PL
,
Weitz
JI
.
Emerging anticoagulant strategies
.
Blood
.
2017
;
129
(
2
):
147
-
154
.
39.
Naudin
C
,
Burillo
E
,
Blankenberg
S
,
Butler
L
,
Renné
T
.
Factor XII contact activation
.
Semin Thromb Hemost
.
2017
;
43
(
8
):
814
-
826
.
40.
Key
NS
.
Epidemiologic and clinical data linking factors XI and XII to thrombosis
.
Hematology Am Soc Hematol Educ Program
.
2014
;
2014
(
1
):
66
-
70
.
41.
Preis
M
,
Hirsch
J
,
Kotler
A
, et al
.
Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events
.
Blood
.
2017
;
129
(
9
):
1210
-
1215
.
42.
Castaldi
PA
,
Larrieu
MJ
,
Caen
J
.
Availability of platelet Factor 3 and activation of factor XII in thrombasthenia
.
Nature
.
1965
;
207
(
995
):
422
-
424
.
43.
Walsh
PN
,
Griffin
JH
.
Contributions of human platelets to the proteolytic activation of blood coagulation factors XII and XI
.
Blood
.
1981
;
57
(
1
):
106
-
118
.
44.
Johne
J
,
Blume
C
,
Benz
PM
, et al
.
Platelets promote coagulation factor XII-mediated proteolytic cascade systems in plasma
.
Biol Chem
.
2006
;
387
(
2
):
173
-
178
.
45.
Bäck
J
,
Sanchez
J
,
Elgue
G
,
Ekdahl
KN
,
Nilsson
B
.
Activated human platelets induce factor XIIa-mediated contact activation
.
Biochem Biophys Res Commun
.
2010
;
391
(
1
):
11
-
17
.
46.
Smith
SA
,
Mutch
NJ
,
Baskar
D
,
Rohloff
P
,
Docampo
R
,
Morrissey
JH
.
Polyphosphate modulates blood coagulation and fibrinolysis
.
Proc Natl Acad Sci U S A
.
2006
;
103
(
4
):
903
-
908
.
47.
Ghosh
S
,
Shukla
D
,
Suman
K
, et al
.
Inositol hexakisphosphate kinase 1 maintains hemostasis in mice by regulating platelet polyphosphate levels
.
Blood
.
2013
;
122
(
8
):
1478
-
1486
.
48.
Van Der Meijden
PE
,
Van Schilfgaarde
M
,
Van Oerle
R
,
Renné
T
,
ten Cate
H
,
Spronk
HM
.
Platelet- and erythrocyte-derived microparticles trigger thrombin generation via factor XIIa
.
J Thromb Haemost
.
2012
;
10
(
7
):
1355
-
1362
.
49.
Morrissey
JH
,
Smith
SA
.
Polyphosphate as modulator of hemostasis, thrombosis, and inflammation
.
J Thromb Haemost
.
2015
;
13
(
suppl 1
):
S92
-
S97
.
50.
Mutch
NJ
.
Polyphosphate as a haemostatic modulator
.
Biochem Soc Trans
.
2016
;
44
(
1
):
18
-
24
.
51.
Smith
SA
,
Choi
SH
,
Davis-Harrison
R
, et al
.
Polyphosphate exerts differential effects on blood clotting, depending on polymer size [published correction appears in Blood. 2011;117(12):3477]
.
Blood
.
2010
;
116
(
20
):
4353
-
4359
.
52.
Puy
C
,
Tucker
EI
,
Wong
ZC
, et al
.
Factor XII promotes blood coagulation independent of factor XI in the presence of long-chain polyphosphates
.
J Thromb Haemost
.
2013
;
11
(
7
):
1341
-
1352
.
53.
Ruiz
FA
,
Lea
CR
,
Oldfield
E
,
Docampo
R
.
Human platelet dense granules contain polyphosphate and are similar to acidocalcisomes of bacteria and unicellular eukaryotes
.
J Biol Chem
.
2004
;
279
(
43
):
44250
-
44257
.
54.
Donovan
AJ
,
Kalkowski
J
,
Smith
SA
,
Morrissey
JH
,
Liu
Y
.
Size-controlled synthesis of granular polyphosphate nanoparticles at physiologic salt concentrations for blood clotting
.
Biomacromolecules
.
2014
;
15
(
11
):
3976
-
3984
.
55.
Labberton
L
,
Kenne
E
,
Long
AT
, et al
.
Neutralizing blood-borne polyphosphate in vivo provides safe thromboprotection
.
Nat Commun
.
2016
;
7
:
12616
.
56.
Verhoef
JJ
,
Barendrecht
AD
,
Nickel
KF
, et al
.
Polyphosphate nanoparticles on the platelet surface trigger contact system activation
.
Blood
.
2017
;
129
(
12
):
1707
-
1717
.
57.
Szymusiak
M
,
Donovan
AJ
,
Smith
SA
, et al
.
Colloidal confinement of polyphosphate on gold nanoparticles robustly activates the contact pathway of blood coagulation
.
Bioconjug Chem
.
2016
;
27
(
1
):
102
-
109
.
58.
Yeon
JH
,
Mazinani
N
,
Schlappi
TS
, et al
.
Localization of short-chain polyphosphate enhances its ability to clot flowing blood plasma
.
Sci Rep
.
2017
;
7
:
42119
.
59.
Labberton
L
,
Long
AT
,
Gendler
SJ
, et al
.
A flow cytometry-based assay for procoagulant platelet polyphosphate [published online ahead of print 2016 November 4]
.
Cytometry B Clin Cytom
.
doi:10.1002/cyto.b.21492
.
60.
Alvarenga
PH
,
Xu
X
,
Oliveira
F
, et al
.
Novel family of insect salivary inhibitors blocks contact pathway activation by binding to polyphosphate, heparin, and dextran sulfate
.
Arterioscler Thromb Vasc Biol
.
2013
;
33
(
12
):
2759
-
2770
.
61.
Jain
S
,
Pitoc
GA
,
Holl
EK
, et al
.
Nucleic acid scavengers inhibit thrombosis without increasing bleeding
.
Proc Natl Acad Sci USA
.
2012
;
109
(
32
):
12938
-
12943
.
62.
Smith
SA
,
Choi
SH
,
Collins
JN
,
Travers
RJ
,
Cooley
BC
,
Morrissey
JH
.
Inhibition of polyphosphate as a novel strategy for preventing thrombosis and inflammation
.
Blood
.
2012
;
120
(
26
):
5103
-
5110
.
63.
Travers
RJ
,
Shenoi
RA
,
Kalathottukaren
MT
,
Kizhakkedathu
JN
,
Morrissey
JH
.
Nontoxic polyphosphate inhibitors reduce thrombosis while sparing hemostasis
.
Blood
.
2014
;
124
(
22
):
3183
-
3190
.
64.
Gajsiewicz
JM
,
Smith
SA
,
Morrissey
JH
.
Polyphosphate and RNA differentially modulate the contact pathway of blood clotting
.
J Biol Chem
.
2017
;
292
(
5
):
1808
-
1814
.
65.
Kannemeier
C
,
Shibamiya
A
,
Nakazawa
F
, et al
.
Extracellular RNA constitutes a natural procoagulant cofactor in blood coagulation
.
Proc Natl Acad Sci USA
.
2007
;
104
(
15
):
6388
-
6393
.
66.
van Montfoort
ML
,
Kuijpers
MJ
,
Knaup
VL
, et al
.
Factor XI regulates pathological thrombus formation on acutely ruptured atherosclerotic plaques
.
Arterioscler Thromb Vasc Biol
.
2014
;
34
(
8
):
1668
-
1673
.
67.
Nickel
KF
,
Ronquist
G
,
Langer
F
, et al
.
The polyphosphate-factor XII pathway drives coagulation in prostate cancer-associated thrombosis
.
Blood
.
2015
;
126
(
11
):
1379
-
1389
.
68.
Nickel
KF
,
Spronk
HM
,
Mutch
NJ
,
Renné
T
.
Time-dependent degradation and tissue factor addition mask the ability of platelet polyphosphates in activating factor XII-mediated coagulation
.
Blood
.
2013
;
122
(
23
):
3847
-
3849
.
69.
Wijeyewickrema
LC
,
Lameignere
E
,
Hor
L
, et al
.
Polyphosphate is a novel cofactor for regulation of complement by a serpin, C1 inhibitor
.
Blood
.
2016
;
128
(
13
):
1766
-
1776
.
70.
Wang
L
,
Fraley
CD
,
Faridi
J
,
Kornberg
A
,
Roth
RA
.
Inorganic polyphosphate stimulates mammalian TOR, a kinase involved in the proliferation of mammary cancer cells
.
Proc Natl Acad Sci USA
.
2003
;
100
(
20
):
11249
-
11254
.
71.
Dinarvand
P
,
Hassanian
SM
,
Qureshi
SH
, et al
.
Polyphosphate amplifies proinflammatory responses of nuclear proteins through interaction with receptor for advanced glycation end products and P2Y1 purinergic receptor
.
Blood
.
2014
;
123
(
6
):
935
-
945
.
72.
Liaw
PC
,
Ito
T
,
Iba
T
,
Thachil
J
,
Zeerleder
S
.
DAMP and DIC: the role of extracellular DNA and DNA-binding proteins in the pathogenesis of DIC
.
Blood Rev
.
2016
;
30
(
4
):
257
-
261
.
73.
Hassanian
SM
,
Dinarvand
P
,
Smith
SA
,
Rezaie
AR
.
Inorganic polyphosphate elicits pro-inflammatory responses through activation of the mammalian target of rapamycin complexes 1 and 2 in vascular endothelial cells
.
J Thromb Haemost
.
2015
;
13
(
5
):
860
-
871
.
74.
Semeraro
F
,
Ammollo
CT
,
Morrissey
JH
, et al
.
Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4
.
Blood
.
2011
;
118
(
7
):
1952
-
1961
.
75.
Han
ED
,
MacFarlane
RC
,
Mulligan
AN
,
Scafidi
J
,
Davis
AE
III
.
Increased vascular permeability in C1 inhibitor-deficient mice mediated by the bradykinin type 2 receptor
.
J Clin Invest
.
2002
;
109
(
8
):
1057
-
1063
.
76.
Oschatz
C
,
Maas
C
,
Lecher
B
, et al
.
Mast cells increase vascular permeability by heparin-initiated bradykinin formation in vivo
.
Immunity
.
2011
;
34
(
2
):
258
-
268
.
77.
Hofman
ZL
,
Relan
A
,
Hack
CE
.
Hereditary angioedema attacks: local swelling at multiple sites
.
Clin Rev Allergy Immunol
.
2016
;
50
(
1
):
34
-
40
.
78.
Hofman
ZL
,
Relan
A
,
Zeerleder
S
,
Drouet
C
,
Zuraw
B
,
Hack
CE
.
Angioedema attacks in patients with hereditary angioedema: local manifestations of a systemic activation process
.
J Allergy Clin Immunol
.
2016
;
138
(
2
):
359
-
366
.
79.
Björkqvist
J
,
de Maat
S
,
Lewandrowski
U
, et al
.
Defective glycosylation of coagulation factor XII underlies hereditary angioedema type III
.
J Clin Invest
.
2015
;
125
(
8
):
3132
-
3146
.
80.
Maas
C
,
Oschatz
C
,
Renné
T
.
The plasma contact system 2.0
.
Semin Thromb Hemost
.
2011
;
37
(
4
):
375
-
381
.
81.
Cichon
S
,
Martin
L
,
Hennies
HC
, et al
.
Increased activity of coagulation factor XII (Hageman factor) causes hereditary angioedema type III
.
Am J Hum Genet
.
2006
;
79
(
6
):
1098
-
1104
.
82.
Deroux
A
,
Boccon-Gibod
I
,
Fain
O
, et al
.
Hereditary angioedema with normal C1 inhibitor and factor XII mutation: a series of 57 patients from the French National Center of Reference for Angioedema
.
Clin Exp Immunol
.
2016
;
185
(
3
):
332
-
337
.
83.
Cugno
M
,
Tedeschi
A
,
Nussberger
J
.
Bradykinin in idiopathic non-histaminergic angioedema
.
Clin Exp Allergy
.
2017
;
47
(
1
):
139
-
140
.
84.
de Maat
S
,
Björkqvist
J
,
Suffritti
C
, et al
.
Plasmin is a natural trigger for bradykinin production in patients with hereditary angioedema with factor XII mutations
.
J Allergy Clin Immunol
.
2016
;
138
(
5
):
1414
-
1423.e9
.
85.
Kaplan
AP
,
Austen
KF
.
A prealbumin activator of prekallikrein. II. Derivation of activators of prekallikrein from active Hageman factor by digestion with plasmin
.
J Exp Med
.
1971
;
133
(
4
):
696
-
712
.
86.
Mitchell
JL
,
Lionikiene
AS
,
Georgiev
G
, et al
.
Polyphosphate colocalizes with factor XII on platelet-bound fibrin and augments its plasminogen activator activity
.
Blood
.
2016
;
128
(
24
):
2834
-
2845
.
87.
Marcos-Contreras
OA
,
Martinez de Lizarrondo
S
,
Bardou
I
, et al
.
Hyperfibrinolysis increases blood-brain barrier permeability by a plasmin- and bradykinin-dependent mechanism
.
Blood
.
2016
;
128
(
20
):
2423
-
2434
.
88.
Cicardi
M
,
Aberer
W
,
Banerji
A
, et al
;
HAWK under the patronage of EAACI (European Academy of Allergy and Clinical Immunology)
.
Classification, diagnosis, and approach to treatment for angioedema: consensus report from the Hereditary Angioedema International Working Group
.
Allergy
.
2014
;
69
(
5
):
602
-
616
.
89.
Simão
F
,
Ustunkaya
T
,
Clermont
AC
,
Feener
EP
.
Plasma kallikrein mediates brain hemorrhage and edema caused by tissue plasminogen activator therapy in mice after stroke
.
Blood
.
2017
;
129
(
16
):
2280
-
2290
.
90.
Bender
L
,
Weidmann
H
,
Rose-John
S
,
Renné
T
,
Long
AT
.
Factor XII-driven inflammatory reactions with implications for anaphylaxis
.
Front Immunol
.
2017
;
8
:
1115
.
91.
Stavrou
EX
,
Fang
C
,
Bane
KL
, et al
.
Factor XII and uPAR upregulate neutrophil functions to influence wound healing
.
J Clin Invest
.
2018
;
128
:
944
-
959
.
Sign in via your Institution