Coagulation factor XII (FXII, Hageman factor, EC = 3.4.21.38) is the zymogen of the serine protease, factor XIIa (FXIIa). FXII is converted to FXIIa through autoactivation induced by “contact” to charged surfaces. FXIIa is of crucial importance for fibrin formation in vitro, but deficiency in the protease is not associated with excessive bleeding. For decades, FXII was considered to have no function for coagulation in vivo. Our laboratory developed the first murine knockout model of FXII. Consistent with their human counterparts, FXII−/− mice have a normal hemostatic capacity. However, thrombus formation in FXII−/− mice is largely defective, and the animals are protected from experimental cerebral ischemia and pulmonary embolism. This murine model has created new interest in FXII because it raises the possibility for safe anticoagulation, which targets thrombosis without influence on hemostasis. We recently have identified platelet polyphosphate (an inorganic polymer) and mast cell heparin as in vivo FXII activators with implications on the initiation of thrombosis and edema during hypersensitivity reactions. Independent of its protease activity, FXII exerts mitogenic activity with implications for angiogenesis. The goal of this review is to summarize the in vivo functions of FXII, with special focus to its functions in thrombosis and vascular biology.

Fibrin formation may be initiated by 2 distinct pathways, either triggered by exposure of blood to a damaged vessel wall (extrinsic) or to blood-borne (intrinsic) factors. The intrinsic pathway of coagulation is initiated by factor XII (FXII, Hageman factor), in a reaction involving high molecular weight kininogen (HK) and plasma kallikrein (PK). These factors are collectively referred to as the plasma contact system.1-6  Contact with negatively charged surfaces induces a conformational change in zymogen FXII resulting in a small amount of active FXII (FXIIa).7  FXIIa cleaves PK to generate active PK, which in turn reciprocally activates FXII.8  FXIIa triggers fibrin formation through activation of factor XI (FXI) and also liberates the inflammatory mediator bradykinin (BK) from HK through cleavage by PK.3  Binding of BK to the kinin B2 receptor (B2R) activates proinflammatory signaling pathways that dilate vessels, induce chemotaxis of neutrophils, and increase vascular permeability.9  Thus, the FXIIa-driven contact system has proinflammatory and procoagulant activities via the kallikrein kinin-system and the intrinsic coagulation pathway, respectively (Figure 1). The serpin C1 esterase inhibitor (C1INH) is the major plasma inhibitor of FXIIa and PK and controls proteolytic activity of the contact system.10  Besides C1INH, antithrombin III (ATIII) and PAI-1 also have FXIIa-blocking activity.11  In vitro, FXIIa triggers activation of the classic complement pathway and initiates the fibrinolytic system via PK-mediated urokinase activation.5  Whether FXIIa has the capacity to trigger activation of the complement and fibrinolytic systems in vivo remains uncertain.

Figure 1

The FXII-driven contact system. Contact with negatively charged surfaces activates coagulation FXII on endothelial cells, leukocytes, bacteria, and thrombocytes and initiates procoagulant and proinflammatory proteolytic reactions. Activated FXII triggers fibrin formation through the FXI-mediated intrinsic pathway of coagulation. Simultaneously, activation of prekallikrein by FXIIa leads to generation of the vasoactive peptide BK by PK-mediated cleavage of HK.

Figure 1

The FXII-driven contact system. Contact with negatively charged surfaces activates coagulation FXII on endothelial cells, leukocytes, bacteria, and thrombocytes and initiates procoagulant and proinflammatory proteolytic reactions. Activated FXII triggers fibrin formation through the FXI-mediated intrinsic pathway of coagulation. Simultaneously, activation of prekallikrein by FXIIa leads to generation of the vasoactive peptide BK by PK-mediated cleavage of HK.

Close modal

The enzymology of the FXII-driven contact system in vitro is well understood. However, its in vivo contributions are just beginning to emerge. FXII-contact activation in vitro provides the mechanistic basis for one of the most commonly used diagnostic coagulation tests, the activated partial thromboplastin time (aPTT), which is extensively used in clinical practice (> 500 million assays/per year worldwide) for preoperative screening, the diagnostics of thrombosis-related autoimmune diseases, and monitoring of anticoagulation therapy. Despite its contribution to fibrin formation in vitro, FXII-initiated coagulation in vivo was not considered to be of significance. This premise is based on the observation that FXII-deficient persons and animals do not exhibit a clinically relevant bleeding phenotype: persons with partial or severe FXII deficiency do not bleed excessively from sites of injury despite a marked prolongation of the aPTT.12,13  This apparent discrepancy between the essential role of FXII for contact-driven fibrin formation in test tubes that eventually lacks a correlation in vivo puzzled investigators for decades. Similar to FXII deficiency, persons lacking the contact proteins PK or HK do not have impaired hemostasis and are commonly diagnosed during routine coagulation screening when a prolonged aPTT is discovered. In contrast, patients deficient in FXI have a mild trauma-induced bleeding disorder (sometimes called “hemophilia C”) that is mostly restricted to tissues with high fibrinolytic activity. Severe FXI deficiency is a rare inherited abnormality in the general population (seen with a 1 in a million people prevalence), but is more common in specific populations, such as Ashkenazi Jews (1 in 450).14 

This lack of a bleeding tendency observed with FXII deficiency is in sharp contrast to deficiencies of other components of the coagulation cascade, such as FVII, tissue factor (TF) and FVIII or FIX (causing the bleeding disorders hemophilia A and B, respectively) and has led to the reasonable hypothesis that fibrin formation in vivo is initiated largely, if not exclusively, through the extrinsic pathway of coagulation. Notably, complete ablation of TF expression causes embryolethal intrauterine bleeding in mice. Human TF deficiency has not been described, indicating that TF is essential for development and/or survival.15  The dominant role of VIIa/TF-driven coagulation for hemostasis is supported by the “revised model of coagulation,” which shows that FXI, the substrate of FXIIa during contact-initiated clotting, can also be activated by thrombin, independently of FXII.16  Although FXII is dispensable for fibrin formation in hemostatic reactions, severe deficiencies (< 10% of normal plasma levels) in the coagulation protein are invariably rare,17  suggesting that FXII has critical roles that warrant future studies. Interestingly, FXII is a rather modern protein in terms of evolution. Copies of the FXII gene are absent in inframammalian vertebrates, such as birds or fish,18  which have a closed circulatory system, supporting the notion that FXII is not required to seal vessel injuries.

Injury to a blood vessel triggers activation of blood platelets and the plasma coagulation system, leading to formation of a blood clot consisting of platelets and fibrin. To investigate the role of FXII in vivo, FXII−/− mice were generated and characterized in experimental thrombosis models.19,20  Identical to FXII-deficient humans, FXII−/− mice have a normal hemostatic capacity as assessed by a tail-bleeding assay and could undergo surgical procedures without excessive bleeding.21  Completely unexpected, intravital fluorescence microscopy and blood flow measurements in 3 distinct arterial beds revealed a severe defect in thrombus formation in FXII-deficient mice when challenged by chemical (FeCl3), mechanical, and Rose Bengal/laser of vascular injury models.21,22  The thrombo-protective phenotype of FXII−/− mice in combination with their normal hemostatic capacity challenges the previously accepted notion of a “coagulation balance.” Although FXII has a crucial role in fibrin formation during “pathologic” thrombosis, it does not contribute to “physiologic” hemostatic fibrin formation at sites of vessel injury. Reconstitution of FXII−/− mice with human FXII normalized both their prolonged aPTT and the defective thrombotic response. The reconstitution experiments indicate that FXII operates similarly in mice and humans. Indeed, the contact system is highly conserved among mammalian species.23  Thrombus formation in FXII heterozygous mice (having 50% of normal FXII plasma levels) was similar to wild-type animals (having 100%), indicating that half of normal plasma concentration is sufficient for vessel-occlusive clot formation. Modulating FXII expression levels by antisense nucleotides showed that reduction of more than 75% FXII antigen plasma levels is required to produce thrombo-protection in a model of stasis-induced venous thrombosis.24 

The decisive role of FXII for experimental models of thrombus formation extends to thromboembolic disease: FXII-deficient mice are protected from cerebral ischemia in an experimental stroke model.25  These thrombo-protective effects are conferred by an impaired FXII-dependent fibrin formation in the microvasculature of the ischemic tissue. Indeed, mice lacking FXI are similarly protected from vessel-occlusive fibrin formation, suggesting that FXII impacts on pathologic clotting predominantly via the intrinsic pathway.26,27  This notion is supported by observations that, in models of lethal pulmonary embolism, survival of mice with combined deficiency in FXII and FXI (FXII−/−/FXI−/−) is similar to animals with deficiency in FXII or FXI alone.28  Given the results from mouse models, the concept that pathologic thrombus formation represents a disequilibrium between the enzymatic reactions that produce a clot at a site of vascular injury probably needs revision. Indeed, pharmacologic inhibition of FXII activity may provide an attractive approach for the management of thrombotic disease. For instance, the peptide-based inhibitor PCK (Phe-Pro-Arg-chloromethylketone) provides protection from cerebral ischemia in wild-type mice without causing excessive bleeding at a site of surgical injury.25  Consistently, a recombinant infestin-4–based inhibitor that specifically targets FXIIa, provides protection from cerebral ischemia in experimental stroke models, albeit it did not reduce the hemostatic capacity of inhibitor-treated mice.29  Cumulatively, the findings in animal models raise the exciting possibility that targeting FXII may offer a safe and powerful strategy for prevention or treatment of pathologic thrombosis without the associated risk of hemorrhage that accompanies currently used anticoagulants.30,31  However, the detailed association of FXII deficiency and risk for thrombosis is probably more complex. The instability of arterial thrombi observed in FXII-deficient mice,21  defective polyphosphate (polyP)–driven fibrin formation in FXII-deficient human and mouse plasma,28,32  as well as case reports of pulmonary emboli seen in FXII-deficient persons33  raises an intriguing hypothesis: FXII deficiency may protect from arterial thrombosis as high shear stress interferes with propagation of unstable thrombi into the vessel lumen. In contrast, larger thrombi may develop in veins under low shear conditions. However, reduced stability of these thrombi increases their risk to embolize.

Analysis of a large registry involving approximate 9000 patients from Austria supports this dual function of FXII in thrombosis. There is an inverted U-shaped association of FXII plasma levels and overall mortality and mortality from cardiovascular disease.34  Clearly more epidemiologic studies are necessary to define the precise consequences of FXII deficiency in arterial and veno-occlusive disease.

FXII activation by nonphysiologic negatively charged surfaces, such as glass, is well established. Various materials have been identified that have the potency of triggering FXII activation.35  Among the best-characterized agents that initiate FXII-dependent clotting are the white clay minerals kaolin and celite (silica-rich compounds), which are commonly used in aPTT coagulation assays. Moreover, kaolin is also used as a hemostatic agent in vivo to terminate blood loss after injury in combat victims. Although the idea of using FXII activators as hemostatic agents for sealing injuries is attractive, exposure of flowing blood to kaolin triggers thromboembolic events and produces excessive heat at the wound site, resulting in additional burn injury and subsequent tissue necrosis.36 

Initiated by the discovery that FXII is crucial for occlusive thrombus formation,21  there was a renewed search for candidate endogenous activators of this protease. RNA that is released from injured cells was shown to activate FXII in plasma. Administration of RNAse (an enzyme that degrades RNA) to mice before experimental challenge has thrombo-protective effects in a FeCl3-induced carotid artery injury model in mice.37  RNA is released from various types of disintegrating cells. The regulatory mechanisms that allow RNA-driven FXII in thrombosis, but not at injury sites at the vessel wall, remain to be identified. However, oligonucleotides, such as RNA, and nucleotide-associated proteins, such as histones,38  contribute to procoagulant states associated with infections and sepsis. FXII binding to collagen is known to enhance coagulation in vitro.39  This interaction is dependent on repetitive negative charge exposed by collagen fibrils.40  When added to platelet-rich plasma, equine type I collagen (Horm) promotes thrombus formation under flow in an FXII-dependent manner.41  In contrast, FXII activity does not contribute to thrombus formation under flow over atherosclerotic plaque material.42  The presence of platelets dramatically enhanced the FXII-dependent procoagulant capacity of collagen,41  suggesting that it also promotes FXII activation indirectly through release of platelet-derived activators.28  In line with this observation, it has been found that activated platelets support FXII activation in a mechanism that is dependent on integrin αIIb-β3 signaling.43  Moreover, activated platelets support activation of the kallikrein-kinin system,44  as well as fibrin formation in an FXII-dependent manner,45  even in the presence of thrombin and TF inhibition.44,46  Taken together, these studies link platelet activation and FXII-driven fibrin formation and raise the question: how do procoagulant platelets activate FXII?

Platelet polyP is an inorganic polymer that is released on platelet activation. Polyphosphate was originally identified in nonmammalian cells and is highly enriched in platelet dense granules.47  Synthetic polyP is used as water softener in technical processes and has been shown to modulate plasma coagulation by multiple mechanisms involving FXII,28  FXI,48  the fibrinolytic system,49  factor V,50  and through alterations of the fibrin structure49  in ex vivo experiments.51  Platelet polyP is a nonbranched polymer of 60-100 orthophosphate residues that directly binds to FXII (and HK) with high affinity.28  Polyphosphate potently initiates coagulation in human plasma and in mice in an FXII-dependent manner. Targeting polyP with phosphatase (an enzyme that cleaves phosphoester bonds and degrades polyP) largely abolishes the formation of occlusive thrombus formation in vivo. Conversely, synthetic polyP restored defective clot formation in platelet-rich plasma from Hermansky-Pudlak patients, who (among others) lack the storage pool for platelet polyP,28  suggesting that polyP may be used as a hemostatic agent. Together, these findings identify polyP as the endogenous platelet-derived FXII activator in vivo.52  While triggering FXII-dependent clotting, polyP/FXII also activates the kallikrein-kinin system leading to PK-mediated BK formation.28  Cumulatively, the identification of polyP as a platelet-derived procoagulant agent provides a long-sought link between primary and secondary hemostasis41  and may represent a new paradigm for the treatment of thromboembolic and inflammatory diseases.53 

TF exposed at subendothelial sites of vessel injury initiates fibrin production. However, tissue factor pathway inhibitor that is released from platelets and endothelial cells rapidly terminates TF activity (Figure 2A). In addition, adherent platelets shield the transmembrane TF protein at the injury site. There is a requirement to stimulate ongoing fibrin production within the 3-dimensionally developing thrombus. polyP released by procoagulant platelets triggers FXII activation and drives fibrin formation via the intrinsic coagulation pathway (Figure 2B). Deficiency in platelet polyP-stimulated FXII-driven fibrin formation, interferes with fibrin fiber thickness,32,49  resulting in mechanical clot instability28  and probably increases the risk of thrombus embolization. Blood-borne TF on circulating monocytes, neutrophils,54  and microparticles,55  which are incorporated into the developing thrombus in a P-selectin–dependent manner,56  provides an alternative FXII-independent mechanism for triggering fibrin production. The relative importance of TF/FVIIa- versus polyP/FXII-initiated fibrin formation and their contribution to thrombosis has not been directly compared and may differ depending on vascular beds and types of vessel injury.

Figure 2

The role of polyP/FXII in thrombosis. (A) Initially, the TF/FVIIa-driven “extrinsic” coagulation pathway triggers fibrin formation at sites of injury. FXII has no function during this stage. Tissue factor pathway inhibitor (TFPI) is released from endothelial cells and adherent platelets and blocks TF activity. (B) In the developing thrombus, activated platelet–released polyP triggers fibrin production via activation of FXII that drives the “intrinsic” coagulation cascade. polyP/FXII-driven fibrin formation operates distant from the injured vessel wall and, hence, does not contribute to hemostasis.

Figure 2

The role of polyP/FXII in thrombosis. (A) Initially, the TF/FVIIa-driven “extrinsic” coagulation pathway triggers fibrin formation at sites of injury. FXII has no function during this stage. Tissue factor pathway inhibitor (TFPI) is released from endothelial cells and adherent platelets and blocks TF activity. (B) In the developing thrombus, activated platelet–released polyP triggers fibrin production via activation of FXII that drives the “intrinsic” coagulation cascade. polyP/FXII-driven fibrin formation operates distant from the injured vessel wall and, hence, does not contribute to hemostasis.

Close modal

Although most investigations focus on FXII as a serine protease, zymogen FXII has mitogenic activities on cultured cells independent of its enzymatic activity. FXII's heavy chain consists of a fibronectin type II and I domain, 2 EGF-like domains, a kringle domain, and a proline-rich region adjacent to its catalytic domain.57  The EGF-like domains in FXII, like those in single-chain urokinase and tissue-type plasminogen activator share structural similarities with homologous domains in hepatocyte growth factor.58  Both FXII and FXIIa are mitogenic on cultured HepG2 cells and phosphorylate MAPK in HepG2 and vascular smooth muscle cells.59  FXII zymogen stimulates 5-bromo-2′-deoxyuridine incorporation through the ERK1/2 and Akt S473 phosphorylation pathway in endothelial cells in a uPAR-dependent manner.60,61  The domain 2 region of uPAR where FXII binds is a regulatory site of the uPAR interactome. In addition to FXII ScuPA, HK/HKa, vitronectin, and PAI-1/uPA, bind to this region to overlapping, but not identical sequences, on uPAR's domain 2 as well. Consistently, FXII stimulates aortic sprouts from wild-type mice but not from uPAR−/− aorta and initiates new vessel formation into Matrigel plugs in wild-type but not in uPAR-deficient animals. Vice versa, there is less number of vessels in skin punch biopsies in a FXII−/− mouse model both constitutively and in a wound healing model.62  In contrast, in another FXII−/− mouse strain, there are no obvious vascular abnormalities in histologic analyses.19 

These combined data indicate that zymogen FXII, like single-chain urokinase, functions as a growth factor that mediates cell signaling leading to proliferation and stimulating angiogenesis, indicating a new in vivo activity for zymogen XII in postnatal angiogenesis after ischemia, inflammation, and injury (Figure 3).

Figure 3

Model of zymogen FXII signaling pathway. FXII binds to domain 2 of uPAR and induces uPAR to communicate intracellularly through β1 integrins. Monoclonal antibody 6S6 to β1 integrin blocks this pathway. Cell stimulation through uPAR and integrin requires an interaction with 1 or more of the ErbB receptor kinases because the tyrosine inhibitors AG1478 or PP3 block FXII signaling. The MEK inhibitor PD98059 blocks FXII-induced ERK1/2 phosphorylation. LY294002, a PI3 kinase inhibitor, blocks FXII-induced Akt phosphorylation. Crosstalk between pERK1/2 and pAkt systems also occurs. Cleaved forms of HK (HKa) block binding of FXII to endothelial cells. Inhibition of any step of the FXII signaling pathways blocks cell proliferation and angiogenesis in HUVEC and aortic segments, respectively. Modified from Falati et al56  with permission.

Figure 3

Model of zymogen FXII signaling pathway. FXII binds to domain 2 of uPAR and induces uPAR to communicate intracellularly through β1 integrins. Monoclonal antibody 6S6 to β1 integrin blocks this pathway. Cell stimulation through uPAR and integrin requires an interaction with 1 or more of the ErbB receptor kinases because the tyrosine inhibitors AG1478 or PP3 block FXII signaling. The MEK inhibitor PD98059 blocks FXII-induced ERK1/2 phosphorylation. LY294002, a PI3 kinase inhibitor, blocks FXII-induced Akt phosphorylation. Crosstalk between pERK1/2 and pAkt systems also occurs. Cleaved forms of HK (HKa) block binding of FXII to endothelial cells. Inhibition of any step of the FXII signaling pathways blocks cell proliferation and angiogenesis in HUVEC and aortic segments, respectively. Modified from Falati et al56  with permission.

Close modal

A severe human FXII deficiency is rare and, as a consequence, there is a lack of epidemiologic studies that systematically compare the incidence or severity of thromboembolic events (ie, stroke, myocardial infarction, pulmonary embolism) in humans with low FXII plasma levels. Based on the animal studies, one would expect that FXII deficiency would protect from thrombosis in patients. In contrast, there is a long history of anecdotal reports suggesting that FXII deficiency may actually predispose to thrombosis.63,64  FXII deficiency associated with increased risk of thrombosis dates to the death of the index patient with FXII deficiency, John Hageman, who died of a pulmonary embolism.65  In brief, railroad worker Hageman fell from a boxcar and fractured his left hemipelvis. He was kept at bed rest for a week and was subsequently allowed to walk on crutches. A few days later, he was found gasping for breath, pulseless, and passed away within minutes. Autopsy showed massive thrombi occluding his left and right pulmonary arteries and several large thrombi were recovered, which presumably had originated from his lower extremity veins.65  It is difficult to link FXII deficiency to this lethal pulmonary embolism event because the trauma and subsequent immobilization represent established FXII-independent risk factors for venous thrombosis. Indeed, careful reanalysis has identified other risk factors in FXII-deficient patients with thrombosis, arguing against FXII deficiency as an independent prothrombotic risk factor.66  Consistently, larger epidemiologic studies in The Netherlands and Switzerland did not find a correlation between FXII deficiency and increased thrombotic risk. However, none of these studies had analyzed whether FXII deficiency conferred thrombo-protection.67,68  In contrast, recent clinical studies from Israel have analyzed the incidence of ischemic stroke and deep vein thrombosis in humans with severe deficiency of FXI (the direct substrate of FXIIa in the intrinsic coagulation pathway). Similarly to FXI-null mice, FXI-deficient humans are largely protected from cerebral ischemia69  and venous thrombosis,70  supporting the decisive role of the intrinsic coagulation pathway for thrombosis in humans.

FXII has the capacity of activating the classic complement pathway in plasma.71  A simultaneous activation of the contact and complement system often occurs in pathologic conditions. Hereditary angioedema (HAE [MIM #106100]) is a life-threatening tissue swelling disorder that develops in persons who are quantitatively or qualitatively deficient in C1-esterase inhibitor (C1INH; HAE type I and II, respectively). C1INH deficiencies facilitate excessive activation of the FXII-driven complement and contact system cascades and the development of edema in HAE type I and II patients.72  In addition to these 2 well-known HAE types, a third variant exists that almost exclusively affects women. HAE type III patients have normal levels of fully functional C1INH but have angioedema nonetheless.73  Clinically, all types of HAE are characterized by recurrent episodes of acute swelling involving the skin, oropharyngeal, laryngeal, or gastrointestinal mucosa. The pathophysiology of the observed increased vascular permeability in HAE has remained controversial. Elegant studies with genetically modified mice demonstrated that edema formation in C1INH-dependent HAE forms is the result of pathologic contact system activation.74  Genetic ablation of C1INH expression results in excessive BK production and excess BK signaling which increases vascular permeability in humans75  and mice.74  In contrast, in combined C1INH and kinin B2 receptor gene-deficient mice vascular leak is normal. Comprehensive studies have identified BK as the principal mediator of vascular leakage in HAE-related swelling attacks in patients.76  Hence, HAE types I and II are treated by infusion of C1INH77  or kinin B2-receptor antagonists (Icatibant).78  Alternatively, recombinant PK inhibitors (Ecallantide) may be used to interfere with acute swelling episodes in HAE patients.79  In contrast to C1INH-dependent forms of HAE, the disease mechanism of HAE type III was enigmatic. Using genome-wide linkage analyses in affected families, HAE type III was shown to be an autosomal dominant disease associated with a single missense mutation (c.1032CrA) in the gene of FXII.80  Consecutive independent studies involving other families found HAE type III to be associated with a different mutation affecting the same nucleotide of the FXII gene, c.1032CrG.81  Both point mutations translate into amino acid exchanges Thr328Lys and Thr328Arg (numbering includes the signal peptide), respectively, on the protein level. The aPTT assay yields normal values in all types of HAE patients and fails to detect affected persons. FXII plasma levels in HAE type III patients are in the normal range,80  suggesting that a yet unknown mechanism triggers edema predominantly in women. HAE patients experience recurrent attacks of swelling, but the stimuli that trigger these periodic episodes of excessive vascular leakage are poorly defined.72 

Until recently, it was thought that mast cell–mediated vascular leakage is predominantly, if not exclusively, mediated by the release of histamine and targeting histamine signaling is widely used therapeutically to treat edema formation associated with aberrant mast cell activity.82  In allergic disease, BK is generated and contributes to increased vascular permeability.78,83,84  Mast cells are highly effective sentinel cells that are found close to blood vessels and are especially common at sites of potential infection.85  A hallmark of mast cell activity in host defense and allergic reactions is increased vascular permeability. In addition to histamine, mast cell secretory granules also contain highly sulfated polysaccharides with heparin as a major constituent. This glycosaminoglycan is synthesized exclusively by mast cells86  and has been identified as a FXII contact activator in vitro.87  Heparin released from allergen-activated mast cells initiates formation of BK in a FXII-dependent manner.88  Minute amounts of heparin (≥ 4μg/mL) are sufficient to activate FXII. FXI is not activated under these circumstances, suggesting the presence of a regulatory mechanism for plasma prekallikrein-directed activity of FXIIa. Heparin also protects FXIIa from inhibition by C1INH.89 

Intravital confocal laser scanning microscopy and tracer extravasation experiments identified BK as the active mediator for increasing leakage in heparin-driven edema in the skin.88  From mast cell–mediated hypersensitivity reactions in mouse models, it was estimated that heparin-driven BK formation accounts for a significant portion (∼ 50%) of total mast-cell evoked increase in vascular permeability. Consistently, small-molecule inhibitors of FXIIa or kinin B2 receptors both interfere with experimental mast cell-triggered leakage. Based on these experimental findings, targeting heparin-initiated BK formation may represent a promising strategy to counteract aberrant mast cell activation in a broad variety of diseases.

The identification of mast cell heparin as an endogenous FXII contact activator in hypersensitivity reactions is reminiscent of reports that had associated therapeutic heparin infusion and contact system activation in a series of life-threatening complications. For decades, heparin has been widely used as an anticoagulant drug. This polysaccharide prevents the formation and extension of blood clots in the circulatory system by increasing AT III activity. Starting November 2007, there was a dramatic increase in heparin-induced adverse reactions in the United States and Germany, such as lethal acute hypersensitivity reactions in patients intravenously receiving commercial heparin of specific lots from a single manufacturer (http://www.fda.gov/cder/drug/infopage/heparin/adverse_events.htm). Consecutively, more than 150 patients died from anaphylactic hypotension associated with intravenous heparin treatment. Comprehensive analyses identified a non-natural contaminant occurring in suspect preparations of heparin that was characterized as oversulfated chondroitin sulfate (OSCS).90  OSCS-contaminated heparin has a greatly increased potency for activating FXII and triggering PK-mediated BK formation in human plasma and in a model of experimental hypotonic shock in vivo.91  These catastrophic reactions in patients are reminiscent of experimental shock models induced by dextran sulfate (DXS)–stimulated BK formation in pigs. Infusion of high-molecular weight DXS (500 kDa) induced transient systemic hypotension,7  and Icatibant (an antagonist of the kinin B2 receptor) blocked this effect on blood pressure.92  The FXII-activating property seems to be dependent on negative charge density of the polysaccharide rather than on a defined structure. Indeed, potency of FXII-driven contact activation in a reconstituted system decreased from dextran sulfate and OSCS (with an average 4 sulfate residues per disaccharide),7,93  to mast cell heparin (with an average of ∼ 2.7 sulfate residues per disaccharide),87  whereas heparan sulfate (with an average of ∼ 1 sulfate residue per disaccharide) was inactive. The potency to activate the plasma contact system also greatly varies among diverse heparin preparations,94  reflecting differences in purification procedures, sources of the polysaccharides, and experimental settings. Of note, although intravenous heparin infusion may trigger BK generation, infusion of the polysaccharide, even at high concentrations in a bolus, normally does not induce hypotension or cause edema. BK that is generated in venous vessels is rapidly and almost completely degraded by angiotensin-converting enzymes and other kininases that are abundantly expressed in lung microvessels before reaching precapillary vascular beds, which regulate blood pressure.

DXS is a polysulfated polysaccharide of linked glucose moiety. DXS-mediated FXII activation is critically dependent on the chain-length and degree of sulfation of the polyanion.7  High molecular weight DXS (500 kDa) is a potent stimulator of FXII activation, whereas shorter DXS polymers fail to independently activate FXII but do support cleavage of FXII by PK.95  Although long-chain DXS induces BK-mediated hypotension in vivo,92  it does not trigger intravascular coagulation. This indicates that some FXII activators do not have the capacity of triggering coagulation and reveal selectivity in the responses to FXII contact-activating surfaces. Indeed, several FXII contact activators initiate unilateral activation of the kallikrein-kinin system. For instance, other polysaccharides besides DXS, such as OSCS or heparin, specifically initiate BK formation without triggering a procoagulant activity. Misfolded protein aggregates, the toxic protein species among others found in the cerebrospinal fluid of Alzheimer disease patients and the plasma of patients with amyloidosis, trigger FXII activation.96  These hazardous protein aggregates specifically initiate BK formation via activation of the kallikrein-kinin system but do not trigger activation of the intrinsic pathway of coagulation. The mechanism for selective activation of PK without activation of homologous FXI97  is not entirely clear but might reflect higher plasma concentrations of PK as of FXI. Furthermore, increased affinity of heparin-dependent plasma inhibitors, such as antithrombin III (AT III) for activated FXI versus PK, might direct FXII activation driven by charged polysaccharides to BK formation.91  Notably, both PK and FXI are surface-bound via HK and share a conserved HK-binding site.97-99  In addition, surface characteristics of the contact activator might be decisive for PK and/or FXI activation. FXII binds differently to negatively charged surfaces, such as polyP, compared with its binding to misfolded protein aggregates. Anionic surface binding is thought to be mediated through the type II fibronectin domain, the second EGF domain, and kringle domain,100,101  whereas interaction with misfolded protein aggregates is mediated by the fibronectin type I domain.102  Differences in FXII binding may also modulate the conversion of α-FXIIa to β-FXIIa by kallikrein activity and shift FXII activity toward BK formation.103,104 

The discovery that pathologic thrombus formation in FXII−/− mice is largely defective in models for experimental arterial thrombosis and ischemic stroke has created a new interest in this protein, especially because it raises the possibility of treating thrombosis without compromising hemostasis. Recently, several in vivo contact activators of FXII have been identified, including platelet polyP, oversulfated chondroitin sulfate, nucleotides, misfolded protein aggregates, and mast cell heparin. These various activators may be involved in the development of thrombotic and/or inflammatory diseases. In addition, investigations on the growth factor function of FXII have only begun. Cumulatively, these discoveries may help elucidate the physiologic function(s) of FXII in vivo, which has remained persistently mysterious since the protein was discovered more than 50 years ago. Further investigations will expectedly reveal novel roles of FXII through which certain disease states, such as thrombosis, inflammation, and infections, can be therapeutically modified.

This work was supported in part by Vetenskapsrådet (K2010-64X-21462-01-3), Hjärt Lungfonden (20090642 and 20110500), Stockholms läns landsting (ALF 20090540 and 2110471), Cancerfonden (100615), and the Federal Ministry of Education and Research (01EO1003 program and the StG-2012-311575; T.R.). A.H.S. was supported by the National Institutes of Health, National Heart, Lung, and Blood Institute (R01 HL052779-15 and R21 HL112666-01). C.M. was supported by The Netherlands Organization for Scientific Research (Veni fellowship 016-126-159).

National Institutes of Health

Contribution: T.R., A.H.S., K.F.N., M.B., and C.M. wrote the review and discussed its contents.

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

Correspondence: Thomas Renné, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna (L1:00), SE-171 76, Stockholm, Sweden; e-mail: thomas@renne.net.

1
Colman
 
RW
Schmaier
 
AH
Contact system: a vascular biology modulator with anticoagulant, profibrinolytic, antiadhesive, and proinflammatory attributes.
Blood
1997
, vol. 
90
 
10
(pg. 
3819
-
3843
)
2
Kaplan
 
AP
Joseph
 
K
Silverberg
 
M
Pathways for bradykinin formation and inflammatory disease.
J Allergy Clin Immunol
2002
, vol. 
109
 
2
(pg. 
195
-
209
)
3
Müller
 
F
Renne
 
T
Novel roles for factor XII-driven plasma contact activation system.
Curr Opin Hematol
2008
, vol. 
15
 
5
(pg. 
516
-
521
)
4
Renné
 
T
The procoagulant and proinflammatory plasma contact system.
Semin Immunopathol
2012
, vol. 
34
 
1
(pg. 
31
-
41
)
5
Saito
 
H
Matsushita
 
T
Kojima
 
T
Historical perspective and future direction of coagulation research.
J Thromb Haemost
2011
, vol. 
9
 
Suppl 1
(pg. 
352
-
363
)
6
Schmaier
 
AH
McCrae
 
KR
The plasma kallikrein-kinin system: its evolution from contact activation.
J Thromb Haemost
2007
, vol. 
5
 
12
(pg. 
2323
-
2329
)
7
Samuel
 
M
Pixley
 
RA
Villanueva
 
MA
Colman
 
RW
Villanueva
 
GB
Human factor XII (Hageman factor) autoactivation by dextran sulfate: circular dichroism, fluorescence, and ultraviolet difference spectroscopic studies.
J Biol Chem
1992
, vol. 
267
 
27
(pg. 
19691
-
19697
)
8
Cochrane
 
CG
Revak
 
SD
Wuepper
 
KD
Activation of Hageman factor in solid and fluid phases: a critical role of kallikrein.
J Exp Med
1973
, vol. 
138
 
6
(pg. 
1564
-
1583
)
9
Leeb-Lundberg
 
LM
Marceau
 
F
Muller-Esterl
 
W
Pettibone
 
DJ
Zuraw
 
BL
Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences.
Pharmacol Rev
2005
, vol. 
57
 
1
(pg. 
27
-
77
)
10
Revak
 
SD
Cochrane
 
CG
The relationship of structure and function in human Hageman factor: the association of enzymatic and binding activities with separate regions of the molecule.
J Clin Invest
1976
, vol. 
57
 
4
(pg. 
852
-
860
)
11
Schapira
 
M
Major inhibitors of the contact phase coagulation factors.
Semin Thromb Hemost
1987
, vol. 
13
 
1
(pg. 
69
-
78
)
12
Lämmle
 
B
Wuillemin
 
W
Huber
 
I
, et al. 
Thrombo-embolism and bleeding tendency in congenital factor XIi deficiency: a study of 74 subjects from 14 Swiss families.
Thromb Haemost
1991
, vol. 
65
 (pg. 
117
-
121
)
13
Ratnoff
 
OD
Colopy
 
JE
A familial hemorrhagic trait associated with a deficiency of a clot-promoting fraction of plasma.
J Clin Invest
1955
, vol. 
34
 
4
(pg. 
602
-
613
)
14
Seligsohn
 
U
Factor XI deficiency in humans.
J Thromb Haemost
2009
, vol. 
7
 
Suppl 1
(pg. 
84
-
87
)
15
Mackman
 
N
Role of tissue factor in hemostasis, thrombosis, and vascular development.
Arterioscler Thromb Vasc Biol
2004
, vol. 
24
 
6
(pg. 
1015
-
1022
)
16
Gailani
 
D
Broze
 
GJ
Factor XI activation in a revised model of blood coagulation.
Science
1991
, vol. 
253
 
5022
(pg. 
909
-
912
)
17
Schloesser
 
M
Zeerleder
 
S
Lutze
 
G
, et al. 
Mutations in the human factor XII gene.
Blood
1997
, vol. 
90
 
10
(pg. 
3967
-
3977
)
18
Ponczek
 
MB
Gailani
 
D
Doolittle
 
RF
Evolution of the contact phase of vertebrate blood coagulation.
J Thromb Haemost
2008
, vol. 
6
 
11
(pg. 
1876
-
1883
)
19
Pauer
 
HU
Renne
 
T
Hemmerlein
 
B
, et al. 
Targeted deletion of murine coagulation factor XII gene: a model for contact phase activation in vivo.
Thromb Haemost
2004
, vol. 
92
 
3
(pg. 
503
-
508
)
20
Iwaki
 
T
Cruz-Topete
 
D
Castellino
 
FJ
A complete factor XII deficiency does not affect coagulopathy, inflammatory responses, and lethality, but attenuates early hypotension in endotoxemic mice.
J Thromb Haemost
2008
, vol. 
6
 
11
(pg. 
1993
-
1995
)
21
Renné
 
T
Pozgajova
 
M
Gruner
 
S
, et al. 
Defective thrombus formation in mice lacking coagulation factor XII.
J Exp Med
2005
, vol. 
202
 
2
(pg. 
271
-
281
)
22
Cheng
 
Q
Tucker
 
EI
Pine
 
MS
, et al. 
A role for factor XIIa-mediated factor XI activation in thrombus formation in vivo.
Blood
2010
, vol. 
116
 
19
(pg. 
3981
-
3989
)
23
Doolittle
 
RF
Coagulation in vertebrates with a focus on evolution and inflammation.
J Innate Immun
2011
, vol. 
3
 
1
(pg. 
9
-
16
)
24
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
, vol. 
118
 
19
(pg. 
5302
-
5311
)
25
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
, vol. 
203
 
3
(pg. 
513
-
518
)
26
Wang
 
X
Smith
 
PL
Hsu
 
MY
, et al. 
Effects of factor XI deficiency on ferric chloride-induced vena cava thrombosis in mice.
J Thromb Haemost
2006
, vol. 
4
 
9
(pg. 
1982
-
1988
)
27
Wang
 
X
Cheng
 
Q
Xu
 
L
, et al. 
Effects of factor IX or factor XI deficiency on ferric chloride-induced carotid artery occlusion in mice.
J Thromb Haemost
2005
, vol. 
3
 
4
(pg. 
695
-
702
)
28
Müller
 
F
Mutch
 
NJ
Schenk
 
WA
, et al. 
Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo.
Cell
2009
, vol. 
139
 
6
(pg. 
1143
-
1156
)
29
Hagedorn
 
I
Schmidbauer
 
S
Pleines
 
I
, et al. 
Factor XIIa inhibitor recombinant human albumin Infestin-4 abolishes occlusive arterial thrombus formation without affecting bleeding.
Circulation
2010
, vol. 
121
 
13
(pg. 
1510
-
1517
)
30
Gailani
 
D
Renne
 
T
The intrinsic pathway of coagulation: a target for treating thromboembolic disease?
J Thromb Haemost
2007
, vol. 
5
 
6
(pg. 
1106
-
1112
)
31
Renné
 
T
Nieswandt
 
B
Gailani
 
D
The intrinsic pathway of coagulation is essential for thrombus stability in mice.
Blood Cells Mol Dis
2006
, vol. 
36
 
2
(pg. 
148
-
151
)
32
Smith
 
SA
Morrissey
 
JH
Polyphosphate enhances fibrin clot structure.
Blood
2008
, vol. 
112
 
7
(pg. 
2810
-
2816
)
33
Mangal
 
AK
Naiman
 
SC
Hageman factor deficiency and oral contraceptives.
Lancet
1980
, vol. 
1
 
8171
pg. 
774
 
34
Endler
 
G
Marsik
 
C
Jilma
 
B
Schickbauer
 
T
Quehenberger
 
P
Mannhalter
 
C
Evidence of a U-shaped association between factor XII activity and overall survival.
J Thromb Haemost
2007
, vol. 
5
 
6
(pg. 
1143
-
1148
)
35
Maas
 
C
Oschatz
 
C
Renne
 
T
The plasma contact system 2.0.
Semin Thromb Hemost
2011
, vol. 
37
 
4
(pg. 
375
-
381
)
36
Kheirabadi
 
BS
Mace
 
JE
Terrazas
 
IB
, et al. 
Clot-inducing minerals versus plasma protein dressing for topical treatment of external bleeding in the presence of coagulopathy.
J Trauma
2010
, vol. 
69
 
5
(pg. 
1062
-
1072
discussion 1072-1073
37
Kannemeier
 
C
Shibamiya
 
A
Nakazawa
 
F
, et al. 
Extracellular RNA constitutes a natural procoagulant cofactor in blood coagulation.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
15
(pg. 
6388
-
6393
)
38
Xu
 
J
Zhang
 
X
Pelayo
 
R
, et al. 
Extracellular histones are major mediators of death in sepsis.
Nat Med
2009
, vol. 
15
 
11
(pg. 
1318
-
1321
)
39
Wilner
 
GD
Nossel
 
HL
LeRoy
 
EC
Activation of Hageman factor by collagen.
J Clin Invest
1968
, vol. 
47
 
12
(pg. 
2608
-
2615
)
40
Nossel
 
HL
Wilner
 
GD
LeRoy
 
EC
Importances of polar groups for initiating blood coagulation and aggregating platelets.
Nature
1969
, vol. 
221
 
5175
(pg. 
75
-
76
)
41
van der Meijden
 
PE
Munnix
 
IC
Auger
 
JM
, et al. 
Dual role of collagen in factor XII-dependent thrombus formation.
Blood
2009
, vol. 
114
 
4
(pg. 
881
-
890
)
42
Reininger
 
AJ
Bernlochner
 
I
Penz
 
SM
, et al. 
A 2-step mechanism of arterial thrombus formation induced by human atherosclerotic plaques.
J Am Coll Cardiol
2010
, vol. 
55
 
11
(pg. 
1147
-
1158
)
43
Castaldi
 
PA
Larrieu
 
MJ
Caen
 
J
Availability of platelet factor 3 and activation of factor XII in thrombasthenia.
Nature
1965
, vol. 
207
 
995
(pg. 
422
-
424
)
44
Johne
 
J
Blume
 
C
Benz
 
PM
, et al. 
Platelets promote coagulation factor XII-mediated proteolytic cascade systems in plasma.
Biol Chem
2006
, vol. 
387
 
2
(pg. 
173
-
178
)
45
Walsh
 
PN
Griffin
 
JH
Contributions of human platelets to the proteolytic activation of blood coagulation factors XII and XI.
Blood
1981
, vol. 
57
 
1
(pg. 
106
-
118
)
46
Bäck
 
J
Sanchez
 
J
Elgue
 
G
Ekdahl
 
KN
Nilsson
 
B
Activated human platelets induce factor XIIa-mediated contact activation.
Biochem Biophys Res Commun
2010
, vol. 
391
 
1
(pg. 
11
-
17
)
47
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
, vol. 
279
 
43
(pg. 
44250
-
44257
)
48
Choi
 
SH
Smith
 
SA
Morrissey
 
JH
Polyphosphate is a cofactor for the activation of factor XI by thrombin.
Blood
2011
, vol. 
118
 
26
(pg. 
6963
-
6970
)
49
Mutch
 
NJ
Engel
 
R
Uitte de Willige
 
S
Philippou
 
H
Ariens
 
RA
Polyphosphate modifies the fibrin network and down-regulates fibrinolysis by attenuating binding of tPA and plasminogen to fibrin.
Blood
2010
, vol. 
115
 
19
(pg. 
3980
-
3988
)
50
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
, vol. 
103
 
4
(pg. 
903
-
908
)
51
Morrissey
 
JH
Choi
 
SH
Smith
 
SA
Polyphosphate: an ancient molecule that links platelets, coagulation, and inflammation.
Blood
2012
, vol. 
119
 
25
(pg. 
5972
-
5979
)
52
Mackman
 
N
Gruber
 
A
Platelet polyphosphate: an endogenous activator of coagulation factor XII.
J Thromb Haemost
2010
, vol. 
8
 
5
(pg. 
865
-
867
)
53
Yun
 
TH
Morrissey
 
JH
Polyphosphate and omptins: novel bacterial procoagulant agents.
J Cell Mol Med
2009
, vol. 
13
 
10
(pg. 
4146
-
4153
)
54
Darbousset
 
R
Thomas
 
GM
Mezouar
 
S
, et al. 
Tissue factor-positive neutrophils bind to injured endothelial wall and initiate thrombus formation.
Blood
2012
, vol. 
120
 
10
(pg. 
2133
-
2143
)
55
Chou
 
J
Mackman
 
N
Merrill-Skoloff
 
G
Pedersen
 
B
Furie
 
BC
Furie
 
B
Hematopoietic cell-derived microparticle tissue factor contributes to fibrin formation during thrombus propagation.
Blood
2004
, vol. 
104
 
10
(pg. 
3190
-
3197
)
56
Falati
 
S
Liu
 
Q
Gross
 
P
, et al. 
Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin.
J Exp Med
2003
, vol. 
197
 
11
(pg. 
1585
-
1598
)
57
Cool
 
DE
Edgell
 
CJ
Louie
 
GV
Zoller
 
MJ
Brayer
 
GD
MacGillivray
 
RT
Characterization of human blood coagulation factor XII cDNA: prediction of the primary structure of factor XII and the tertiary structure of beta-factor XIIa.
J Biol Chem
1985
, vol. 
260
 
25
(pg. 
13666
-
13676
)
58
Miyazawa
 
K
Shimomura
 
T
Kitamura
 
A
Kondo
 
J
Morimoto
 
Y
Kitamura
 
N
Molecular cloning and sequence analysis of the cDNA for a human serine protease responsible for activation of hepatocyte growth factor: structural similarity of the protease precursor to blood coagulation factor XII.
J Biol Chem
1993
, vol. 
268
 
14
(pg. 
10024
-
10028
)
59
Schmeidler-Sapiro
 
KT
Ratnoff
 
OD
Gordon
 
EM
Mitogenic effects of coagulation factor XII and factor XIIa on HepG2 cells.
Proc Natl Acad Sci U S A
1991
, vol. 
88
 
10
(pg. 
4382
-
4385
)
60
Fernando
 
LP
Natesan
 
S
Joseph
 
K
Kaplan
 
AP
High molecular weight kininogen and factor XII binding to endothelial cells and astrocytes.
Thromb Haemost
2003
, vol. 
90
 
5
(pg. 
787
-
795
)
61
Gordon
 
EM
Venkatesan
 
N
Salazar
 
R
, et al. 
Factor XII-induced mitogenesis is mediated via a distinct signal transduction pathway that activates a mitogen-activated protein kinase.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 
5
(pg. 
2174
-
2179
)
62
LaRusch
 
GA
Mahdi
 
F
Shariat-Madar
 
Z
, et al. 
Factor XII stimulates ERK1/2 and Akt through uPAR, integrins, and the EGFR to initiate angiogenesis.
Blood
2010
, vol. 
115
 
24
(pg. 
5111
-
5120
)
63
Bach
 
J
Endler
 
G
Winkelmann
 
BR
, et al. 
Coagulation factor XII (FXII) activity, activated FXII, distribution of FXII C46T gene polymorphism and coronary risk.
J Thromb Haemost
2008
, vol. 
6
 
2
(pg. 
291
-
296
)
64
Halbmayer
 
WM
Mannhaltaer
 
C
Feichtinger
 
C
Rubi
 
K
Fischer
 
M
The prevalence of factor XII deficiency in 103 orally anticoagulated outpatients suffering from recurrent venous and/or arterial thromboembolism.
Thromb Haemost
1992
, vol. 
68
 pg. 
285
 
65
Ratnoff
 
OD
The demise of John Hageman.
N Engl J Med
1968
, vol. 
279
 (pg. 
760
-
761
)
66
Girolami
 
A
Randi
 
ML
Gavasso
 
S
Lombardi
 
AM
Spiezia
 
F
The occasional venous thromboses seen in patients with severe (homozygous) FXII deficiency are probably due to associated risk factors: a study of prevalence in 21 patients and review of the literature.
J Thromb Thrombolysis
2004
, vol. 
17
 
2
(pg. 
139
-
143
)
67
Zeerleder
 
S
Schloesser
 
M
Redondo
 
M
, et al. 
Reevaluation of the incidence of thromboembolic complications in congenital factor XII deficiency: a study on 73 subjects from 14 Swiss families.
Thromb Haemost
1999
, vol. 
82
 
4
(pg. 
1240
-
1246
)
68
Koster
 
T
Rosendaal
 
FR
Briet
 
E
Vandenbroucke
 
JP
John Hageman's factor and deep-vein thrombosis: Leiden Thrombophilia Study.
Br J Haematol
1994
, vol. 
87
 
2
(pg. 
422
-
424
)
69
Salomon
 
O
Steinberg
 
DM
Koren-Morag
 
N
Tanne
 
D
Seligsohn
 
U
Reduced incidence of ischemic stroke in patients with severe factor XI deficiency.
Blood
2008
, vol. 
111
 
8
(pg. 
4113
-
4117
)
70
Salomon
 
O
Steinberg
 
DM
Zucker
 
M
Varon
 
D
Zivelin
 
A
Seligsohn
 
U
Patients with severe factor XI deficiency have a reduced incidence of deep-vein thrombosis.
Thromb Haemost
2011
, vol. 
105
 
2
(pg. 
269
-
273
)
71
Ghebrehiwet
 
B
Silverberg
 
M
Kaplan
 
AP
Activation of the classical pathway of complement by Hageman factor fragment.
J Exp Med
1981
, vol. 
153
 
3
(pg. 
665
-
676
)
72
Zuraw
 
BL
Clinical practice: hereditary angioedema.
N Engl J Med
2008
, vol. 
359
 
10
(pg. 
1027
-
1036
)
73
Bork
 
K
Barnstedt
 
SE
Koch
 
P
Traupe
 
H
Hereditary angioedema with normal C1-inhibitor activity in women.
Lancet
2000
, vol. 
356
 
9225
(pg. 
213
-
217
)
74
Han
 
ED
MacFarlane
 
RC
Mulligan
 
AN
Scafidi
 
J
Davis
 
AE
Increased vascular permeability in C1 inhibitor-deficient mice mediated by the bradykinin type 2 receptor.
J Clin Invest
2002
, vol. 
109
 
8
(pg. 
1057
-
1063
)
75
Cugno
 
M
Cicardi
 
M
Bottasso
 
B
, et al. 
Activation of the coagulation cascade in C1-inhibitor deficiencies.
Blood
1997
, vol. 
89
 
9
(pg. 
3213
-
3218
)
76
Nussberger
 
J
Cugno
 
M
Cicardi
 
M
Bradykinin-mediated angioedema.
N Engl J Med
2002
, vol. 
347
 
8
(pg. 
621
-
622
)
77
Zuraw
 
BL
Busse
 
PJ
White
 
M
, et al. 
Nanofiltered C1 inhibitor concentrate for treatment of hereditary angioedema.
N Engl J Med
2010
, vol. 
363
 
6
(pg. 
513
-
522
)
78
Cicardi
 
M
Banerji
 
A
Bracho
 
F
, et al. 
Icatibant, a new bradykinin-receptor antagonist, in hereditary angioedema.
N Engl J Med
2010
, vol. 
363
 
6
(pg. 
532
-
541
)
79
Cicardi
 
M
Levy
 
RJ
McNeil
 
DL
, et al. 
Ecallantide for the treatment of acute attacks in hereditary angioedema.
N Engl J Med
2010
, vol. 
363
 
6
(pg. 
523
-
531
)
80
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
, vol. 
79
 
6
(pg. 
1098
-
1104
)
81
Dewald
 
G
Bork
 
K
Missense mutations in the coagulation factor XII (Hageman factor) gene in hereditary angioedema with normal C1 inhibitor.
Biochem Biophys Res Commun
2006
, vol. 
343
 
4
(pg. 
1286
-
1289
)
82
Galli
 
SJ
Grimbaldeston
 
M
Tsai
 
M
Immunomodulatory mast cells: negative, as well as positive, regulators of immunity.
Nat Rev Immunol
2008
, vol. 
8
 
6
(pg. 
478
-
486
)
83
Proud
 
D
Kaplan
 
AP
Kinin formation: mechanisms and role in inflammatory disorders.
Annu Rev Immunol
1988
, vol. 
6
 (pg. 
49
-
83
)
84
Proud
 
D
Togias
 
A
Naclerio
 
RM
Crush
 
SA
Norman
 
PS
Lichtenstein
 
LM
Kinins are generated in vivo following nasal airway challenge of allergic individuals with allergen.
J Clin Invest
1983
, vol. 
72
 
5
(pg. 
1678
-
1685
)
85
Marshall
 
JS
Mast-cell responses to pathogens.
Nat Rev Immunol
2004
, vol. 
4
 
10
(pg. 
787
-
799
)
86
Forsberg
 
E
Pejler
 
G
Ringvall
 
M
, et al. 
Abnormal mast cells in mice deficient in a heparin-synthesizing enzyme.
Nature
1999
, vol. 
400
 
6746
(pg. 
773
-
776
)
87
Hojima
 
Y
Cochrane
 
CG
Wiggins
 
RC
Austen
 
KF
Stevens
 
RL
In vitro activation of the contact (Hageman factor) system of plasma by heparin and chondroitin sulfate E.
Blood
1984
, vol. 
63
 
6
(pg. 
1453
-
1459
)
88
Oschatz
 
C
Maas
 
C
Lecher
 
B
, et al. 
Mast cells increase vascular permeability by heparin-initiated bradykinin formation in vivo.
Immunity
2011
, vol. 
34
 
2
(pg. 
258
-
268
)
89
Pixley
 
RA
Schmaier
 
A
Colman
 
RW
Effect of negatively charged activating compounds on inactivation of factor XIIa by Cl inhibitor.
Arch Biochem Biophys
1987
, vol. 
256
 
2
(pg. 
490
-
498
)
90
Guerrini
 
M
Beccati
 
D
Shriver
 
Z
, et al. 
Oversulfated chondroitin sulfate is a contaminant in heparin associated with adverse clinical events.
Nat Biotechnol
2008
, vol. 
26
 
6
(pg. 
669
-
675
)
91
Kishimoto
 
TK
Viswanathan
 
K
Ganguly
 
T
, et al. 
Contaminated heparin associated with adverse clinical events and activation of the contact system.
N Engl J Med
2008
, vol. 
358
 
23
(pg. 
2457
-
2467
)
92
Siebeck
 
M
Cheronis
 
JC
Fink
 
E
, et al. 
Dextran sulfate activates contact system and mediates arterial hypotension via B2 kinin receptors.
J Appl Physiol
1994
, vol. 
77
 
6
(pg. 
2675
-
2680
)
93
Schwartz
 
LB
Heparin comes clean.
N Engl J Med
2008
, vol. 
358
 
23
(pg. 
2505
-
2509
)
94
Brunnée
 
T
Reddigari
 
SR
Shibayama
 
Y
Kaplan
 
AP
Silverberg
 
M
Mast cell derived heparin activates the contact system: a link to kinin generation in allergic reactions.
Clin Exp Allergy
1997
, vol. 
27
 
6
(pg. 
653
-
663
)
95
Citarella
 
F
Wuillemin
 
WA
Lubbers
 
YT
Hack
 
CE
Initiation of contact system activation in plasma is dependent on factor XII autoactivation and not on enhanced susceptibility of factor XII for kallikrein cleavage.
Br J Haematol
1997
, vol. 
99
 
1
(pg. 
197
-
205
)
96
Maas
 
C
Govers-Riemslag
 
JW
Bouma
 
B
, et al. 
Misfolded proteins activate factor XII in humans, leading to kallikrein formation without initiating coagulation.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3208
-
3218
)
97
Renné
 
T
Gailani
 
D
Meijers
 
JC
Muller-Esterl
 
W
Characterization of the H-kininogen-binding site on factor XI: a comparison of factor XI and plasma prekallikrein.
J Biol Chem
2002
, vol. 
277
 
7
(pg. 
4892
-
4899
)
98
Herwald
 
H
Renne
 
T
Meijers
 
JC
, et al. 
Mapping of the discontinuous kininogen binding site of prekallikrein: a distal binding segment is located in the heavy chain domain A4.
J Biol Chem
1996
, vol. 
271
 
22
(pg. 
13061
-
13067
)
99
Renné
 
T
Dedio
 
J
Meijers
 
JC
Chung
 
D
Muller-Esterl
 
W
Mapping of the discontinuous H-kininogen binding site of plasma prekallikrein: evidence for a critical role of apple domain-2.
J Biol Chem
1999
, vol. 
274
 
36
(pg. 
25777
-
25784
)
100
Citarella
 
F
Ravon
 
DM
Pascucci
 
B
Felici
 
A
Fantoni
 
A
Hack
 
CE
Structure/function analysis of human factor XII using recombinant deletion mutants: evidence for an additional region involved in the binding to negatively charged surfaces.
Eur J Biochem
1996
, vol. 
238
 
1
(pg. 
240
-
249
)
101
Citarella
 
F
te Velthuis
 
H
Helmer-Citterich
 
M
Hack
 
CE
Identification of a putative binding site for negatively charged surfaces in the fibronectin type II domain of human factor XII: an immunochemical and homology modeling approach.
Thromb Haemost
2000
, vol. 
84
 
6
(pg. 
1057
-
1065
)
102
Maas
 
C
Schiks
 
B
Strangi
 
RD
, et al. 
Identification of fibronectin type I domains as amyloid-binding modules on tissue-type plasminogen activator and three homologs.
Amyloid
2008
, vol. 
15
 
3
(pg. 
166
-
180
)
103
Gebbink
 
MF
Bouma
 
B
Maas
 
C
Bouma
 
BN
Physiological responses to protein aggregates: fibrinolysis, coagulation and inflammation (new roles for old factors).
FEBS Lett
2009
, vol. 
583
 
16
(pg. 
2691
-
2699
)
104
Schmaier
 
AH
The elusive physiologic role of Factor XII.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3006
-
3009
)
Sign in via your Institution