Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Ann Biol Clin (Paris) ; 82(1): 9-23, 2024 04 19.
Article Fr | MEDLINE | ID: mdl-38638015

Thrombosis remains one of the leading causes of death in the world. The history of anticoagulation has evolved considerably from non-specific drugs (i.e., heparins and vitamin K antagonists, VKA) to agents that directly target specific coagulation factors (i.e., argatroban, fondaparinux and direct oral anticoagulants, DOAC). Since the last decade, DOAC are widely used in clinical practice because of their ease to use, their favorable pharmacological profile and the fact that they do not require monitoring. However, despite having a better safety profile than vitamin K antagonist, their bleeding risk is not negligible. New anticoagulants targeting the contact phase of coagulation are currently being developed and could make it possible to prevent the risk of thrombosis without impairing hemostasis. Epidemiological and preclinical data on FXI deficiency make FXI a promising therapeutic target. The aim of this review is to summarize the results of the various clinical trials available that focus on FXI/FXIa inhibition, and to highlight the challenges that this new therapeutic class of anticoagulants will face.


Anticoagulants , Thrombosis , Humans , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Factor XI/pharmacology , Factor XI/therapeutic use , Blood Coagulation/physiology , Thrombosis/drug therapy , Thrombosis/prevention & control , Vitamin K/therapeutic use
2.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(1): 16-22, 2024 Jan.
Article Zh | MEDLINE | ID: mdl-38404266

Anticoagulation therapy stands as a key treatment for thrombotic diseases. The consequential bleeding risk tied to existing anticoagulation methods significantly impacts patient prognosis. In the intensive care unit (ICU), patients often necessitate organ support, leading to the inevitable placement of artificial devices in blood vessels, thereby requiring anticoagulation treatment to avert clot formation that might impede organ support. Nevertheless, these patients commonly encounter a heightened risk of bleeding. Hemophilia B, identified in 1953, manifests as a deficiency in coagulation factor XI (FXI), which focused people's perspective on the endogenous coagulation pathway, that is, the contact pathway. Upon interaction between the surface of artificial devices and FXII, FXII activates, subsequently triggering FXI and initiating the "coagulation cascade" within the contact pathway. Inhibitors targeting the contact pathway encompass two primary categories: FXII inhibitors and FXI inhibitors, capable of impeding this process. This article reviews the role of FXII and FXI in activating the contact pathway, seeking to illuminate their contributions to thrombus formation. By listing the relatively mature drugs and their indications, clinicians are familiar with this new anticoagulant.


Factor XII , Thrombosis , Humans , Factor XII/metabolism , Factor XII/pharmacology , Blood Coagulation , Factor XI/metabolism , Factor XI/pharmacology , Thrombosis/drug therapy , Anticoagulants/therapeutic use
5.
Blood Rev ; 62: 101119, 2023 11.
Article En | MEDLINE | ID: mdl-37580207

Anticoagulant drugs that are currently used to prevent and/or treat thrombosis have some limitations that hinder their ability to meet specific clinical requirements. While these drugs effectively reduce the rates of thrombotic events, they simultaneously increase the risk of bleeding. Moreover, their risk-to-benefit balance is problematic in some patients, such as those with severe chronic kidney disease or those at high bleeding risk. A novel anticoagulation method, FXI inhibition has emerged as a promising alternative. It demonstrates a strong rationale for the prevention and treatment of venous thromboembolism and the potential fulfillment of unmet clinical needs in the cardiovascular field. A number of FXI inhibitors are currently undergoing clinical investigation. The objective of this review is to provide an overview of early results of research on FXI inhibitors in the cardiovascular setting, offering valuable insights into their potential role in shaping the future of anticoagulation.


Cardiovascular Diseases , Thrombosis , Humans , Factor XI/pharmacology , Factor XI/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Blood Coagulation , Thrombosis/drug therapy , Thrombosis/etiology , Thrombosis/prevention & control , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Hemorrhage/etiology , Hemorrhage/prevention & control , Hemorrhage/drug therapy
7.
Eur Heart J ; 44(4): 280-292, 2023 Jan 21.
Article En | MEDLINE | ID: mdl-36263776

Anticoagulants are the cornerstone for prevention and treatment of thrombosis but are not completely effective, and concerns about the risk of bleeding continue to limit their uptake. Animal studies and experience from patients with genetic coagulation factor XI deficiency suggesting that this factor is more important for thrombosis than for haemostasis raises the potential for drugs that target factor XI to provide safer anticoagulation. Multiple factor XI inhibitors are currently under evaluation in clinical trials, including parenterally administered antisense oligonucleotides, monoclonal antibodies, and orally active small-molecule inhibitors. Promising results of phase 2 trials in patients undergoing major orthopaedic surgery, and in those with end-stage kidney disease, atrial fibrillation and acute coronary syndromes have led to large phase 3 trials that are currently ongoing. We here review premises for the use of these agents, results so far accrued, ongoing studies, and perspectives for future patient care.


Factor XI , Thrombosis , Animals , Anticoagulants/therapeutic use , Blood Coagulation , Factor XI/genetics , Factor XI/pharmacology , Factor XI/therapeutic use , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Thrombosis/prevention & control , Thrombosis/drug therapy , Humans
9.
J Cardiovasc Pharmacol ; 79(6): 887-895, 2022 06 01.
Article En | MEDLINE | ID: mdl-35353073

ABSTRACT: Circular RNAs (circRNAs) have been verified as vital regulators in various diseases, including membranous nephropathy (MN). Therefore, the role of circ_CDYL in podocyte apoptosis and MN was investigated. The real-time quantitative polymerase chain reaction was performed to measure the expression of circ_CDYL, microRNA-149-5p (miR-149-5p), and tumor necrosis factor superfamily member 11 (TNFSF11) in podocytes. In addition, angiotensin II (Ang II) was used to induce apoptosis of podocytes. The apoptosis-related protein expression was quantified by western blot assay. The apoptosis of podocytes was evaluated by flow cytometry assay. The interaction relationship between miR-149-5p and circ_CDYL or TNFSF11 was confirmed by dual-luciferase reporter assay. Circ_CDYL was significantly overexpressed in MN patients and Ang II-induced podocytes compared with control groups. Importantly, loss-of-functional experiments indicated that knockdown of circ_CDYL protected podocytes from Ang II-induced apoptosis. MiR-149-5p was verified as target of circ_CDYL and negatively correlated with circ_CDYL expression in MN patients. Knockdown of circ_CDYL-mediated effects on Ang II-induced podocyte cells were abolished by silencing miR-149-5p. Besides, the upregulation of miR-149-5p could suppress apoptosis in Ang II-induced podocyte cells by targeting TNFSF11. Under Ang II stimulation, the upregulation of TNFSF11 could increase the expression of TNFSF11 and induce apoptosis in circ_CDYL-silencing podocytes. Our results confirmed that circ_CDYL specifically targeted miR-149-5p/TNFSF11 pathway to regulate Ang II-induced apoptosis in podocytes, which might be useful diagnostic biomarkers in MN.


Glomerulonephritis, Membranous , MicroRNAs , Podocytes , Angiotensin II/metabolism , Apoptosis , Co-Repressor Proteins/metabolism , Factor XI/metabolism , Factor XI/pharmacology , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Humans , Hydro-Lyases/metabolism , Hydro-Lyases/pharmacology , MicroRNAs/metabolism , Podocytes/metabolism , Podocytes/pathology , RANK Ligand/metabolism
10.
Hamostaseologie ; 41(2): 104-110, 2021 Apr.
Article En | MEDLINE | ID: mdl-33860518

Despite advances in anticoagulant therapy, thrombosis remains the leading cause of morbidity and mortality worldwide. Heparin and vitamin K antagonists (VKAs), the first anticoagulants to be used successfully for the prevention and treatment of thrombosis, are associated with a risk of bleeding. These agents target multiple coagulation factors. Thus, by activating antithrombin, heparin mainly inhibits factor Xa and thrombin, whereas VKAs lower the levels of the vitamin K-dependent clotting factors. Direct oral anticoagulants, which have replaced VKAs for many indications, inhibit only factor Xa or thrombin. Although the direct oral anticoagulants are associated with less bleeding than VKAs, bleeding remains their major side effect. Epidemiological and animal studies have identified factor XI as a target for potentially safer anticoagulant drugs because factor XI deficiency or inhibition protects against thrombosis and is associated with little or no bleeding. Several factor XI-directed strategies are currently under investigation. This article (1) reviews the rationale for the development of factor XI inhibitors, (2) identifies the agents in most advanced stages of development, (3) describes the results of completed clinical trials and provides a summary of those underway, and (4) highlights the opportunities and challenges for this next generation of anticoagulants.


Anticoagulants/therapeutic use , Factor XI/therapeutic use , Thrombosis/drug therapy , Anticoagulants/pharmacology , Factor XI/pharmacology , Humans
11.
Thromb Haemost ; 110(1): 62-75, 2013 Jul.
Article En | MEDLINE | ID: mdl-23636277

Elevated plasma levels of factor VIII (FVIII) are associated with increased risk of deep venous thrombosis. The aim of this study is to elucidate how elevated FVIII levels affect venous thrombus formation and propagation in vivo. We examined rabbit plasma FVIII activity, plasma thrombin generation, whole blood coagulation, platelet aggregation and venous wall thrombogenicity before and one hour after an intravenous infusion of recombinant human FVIII (rFVIII). Venous thrombus induced by the endothelial denudation of rabbit jugular veins was histologically assessed. Thrombus propagation was evaluated as indocyanine green fluorescence intensity. Argatroban, a thrombin inhibitor, and neutralised antibodies for tissue factor (TF), factor XI (FXI), and von Willebrand factor (VWF) were infused before or after thrombus induction to investigate their effects on venous thrombus formation or propagation. Recombinant FVIII (100 IU/kg) increased rabbit plasma FVIII activity two-fold and significantly enhanced whole blood coagulation and total plasma thrombin generation, but did not affect initial thrombin generation time, platelet aggregation and venous wall thrombogenicity. The rFVIII infusion also increased the size of venous thrombus 1 hour after thrombus induction. Argatroban and the antibodies for TF, FXI or VWF inhibited such enhanced thrombus formation and all except TF suppressed thrombus propagation. In conclusion, elevated plasma FVIII levels enhance venous thrombus formation and propagation. Excess thrombin generation by FXI and VWF-mediated FVIII recruitment appear to contribute to the growth of FVIII-driven venous thrombus.


Blood Platelets/metabolism , Factor VIII/metabolism , Factor XI/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Venous Thrombosis/metabolism , von Willebrand Factor/metabolism , Animals , Antibodies, Blocking/administration & dosage , Arginine/analogs & derivatives , Blood Platelets/pathology , Factor VIII/pharmacology , Factor XI/pharmacology , Hemostasis/drug effects , Humans , Jugular Veins/pathology , Jugular Veins/surgery , Male , Pipecolic Acids/administration & dosage , Platelet Aggregation/drug effects , Rabbits , Sulfonamides , Thromboplastin/immunology , Venous Thrombosis/blood , Venous Thrombosis/drug therapy , von Willebrand Factor/pharmacology
12.
Blood Coagul Fibrinolysis ; 17(4): 251-7, 2006 Jun.
Article En | MEDLINE | ID: mdl-16651866

In-vitro studies have shown that thrombin-mediated factor XI activation enhances thrombin and fibrin formation, rendering the clot more thrombogenic and protecting it from lysis by activation of thrombin activatable fibrinolysis inhibitor. These effects of factor XI are only observed when coagulation is initiated by a low concentration of soluble tissue factor. At high concentrations of soluble tissue factor no effects of factor XI are seen on coagulation and fibrinolysis. In vivo, tissue factor is present in large amounts in the vascular wall. This makes it difficult to extrapolate these in-vitro findings on factor XI to the in-vivo situation. To address the question of whether factor XI could play a role in coagulation initiated on a tissue factor-containing surface we devised a static in-vitro coagulation model in which clotting is initiated in recalcified citrated plasma by tissue factor coated on the bottom of microtiter plates. The effect of factor XI was studied with an antibody that blocked the activation of factor IX by activated factor XI. The tissue factor coating strategy produced clotting times similar to those obtained with cultured tissue factor-expressing vessel wall cells (smooth muscle cells, fibroblasts and activated endothelial cells) grown to confluence in the same wells. A factor XI-dependent effect on clot formation and clot lysis was observed depending on the plasma volume used. In clots formed from small amounts of plasma (100 microl) no effect of factor XI was detected. In larger clots (200-300 microl) factor XI not only increased prothrombin activation and the fibrin formation rate but also inhibited fibrinolysis. Effects of factor XI were observed at short clotting times (3-4 min) similar to the clotting times found on cultured tissue factor-expressing vessel wall cells. This is in contrast with earlier studies using soluble tissue factor, in which effects of factor XI were only observed at much longer clotting times using low soluble tissue factor concentrations. We conclude that factor XI not only enhances coagulation initiated by surface bound tissue factor but also protects the clot against lysis once it is formed. On the basis of these results, we propose a coagulation model in which initial clot formation in the proximity of the tissue factor surface is not factor XI dependent. Clot formation becomes dependent on factor XI in the propagation phase when the clot is increasing in size. These findings support a role for factor XI in the propagation of clot growth after tissue factor-dependent initiation.


Factor XI/pharmacology , Fibrin/drug effects , Fibrinolysis/drug effects , Models, Biological , Prothrombin/metabolism , Blood Coagulation/drug effects , Fibrin/biosynthesis , Humans , Surface Properties , Thrombin/biosynthesis , Time Factors
13.
J Biol Chem ; 278(23): 20618-23, 2003 Jun 06.
Article En | MEDLINE | ID: mdl-12663668

Previous studies on the interaction of high molecular weight kininogen (HK) with endothelial cells have reported a large number of binding sites (106-107 sites/cell) with differing relative affinities (KD = 7-130 nm) and have implicated various receptors or receptor complexes. In this study, we examined the binding of HK to human umbilical vein endothelial cells (HUVEC) with a novel assay system utilizing HUVEC immobilized on microcarrier beads, which eliminates the detection of the high affinity binding sites found nonspecifically in conventional microtiter well assays. We report that HK binds to 8.5 x 104 high affinity (KD = 21 nm) sites per HUVEC, i.e. 10-100-fold fewer than previously reported. Although HK binding is unaffected by the presence of a physiological concentration of prekallikrein, factor XI abrogates HK binding to HUVEC in a concentration-dependent manner. Disruption of the naturally occurring complex between factor XI and HK by the addition of a 31-amino acid peptide mimicking the factor XI-binding site on HK restored HK binding to HUVEC. Furthermore, HK inhibited thrombin-stimulated von Willebrand factor release by HUVEC but not thrombin receptor activation peptide (SFLLRN-amide)-stimulated von Willebrand factor release. Factor XI restored the ability of thrombin to stimulate von Willebrand factor release in the presence of low HK concentrations. These results suggest that free HK, or HK in complex with prekallikrein but not in complex with factor XI, interacts with the endothelium and can maintain endothelial cell quiescence by preventing endothelial stimulation by thrombin.


Endothelium, Vascular/metabolism , Factor XI/pharmacology , Kininogen, High-Molecular-Weight/pharmacology , Prekallikrein/pharmacology , Cells, Cultured , Cells, Immobilized , Endothelium, Vascular/cytology , Hemostatics/pharmacology , Humans , Iodine Radioisotopes , Kininogen, High-Molecular-Weight/metabolism , Microspheres , Protein Binding/drug effects , Thrombin/pharmacology , Titrimetry , Umbilical Veins/cytology , von Willebrand Factor/metabolism
14.
Thromb Haemost ; 85(6): 1060-5, 2001 Jun.
Article En | MEDLINE | ID: mdl-11434685

Thrombin generation has been studied in the plasma of severely factor XI deficient patients under conditions in which contact activation did not play a role. In platelet-rich as well as platelet-poor plasma, thrombin generation was dependent upon the presence of factor XI at tissue factor concentrations of between 1 and 20 pg/ml i.e. approximately 0.01 to 0.20% of the concentration normally present in the thromboplastin time determination. The requirement for factor XI is low; significant thrombin generation was seen at 1% factor XI; at 10%, thrombin formation was nearly normalised. A suspension of normal platelets in severely factor XI deficient plasma did not increase thrombin generation. This implies that there is no significant factor XI activity carried by normal platelets, although the presence of factor XI and factor XI inhibitors in platelets cannot be ruled out.


Factor XI/physiology , Thrombin/biosynthesis , Thromboplastin/metabolism , Blood Platelets , Dose-Response Relationship, Drug , Factor XI/pharmacology , Factor XI Deficiency/blood , Factor XI Deficiency/congenital , Family Health , Humans , Kinetics , Thrombin/drug effects , Thromboplastin/pharmacology
15.
Thromb Haemost ; 85(6): 1055-9, 2001 Jun.
Article En | MEDLINE | ID: mdl-11434684

Blood coagulation has been thought to be composed of both intrinsic and extrinsic pathways. Recent evidence strongly supports the critical role of the extrinsic pathway in the initiation of blood coagulation. This investigation established an assay that examines the role of FXI in the thromboplastin-initiated (extrinsic) coagulation based on this new concept. Plasma clotting times were measured at different concentrations of thromboplastin with activated FXII inhibited (FXIIa-inhibited Diluted Thromboplastin Time, FXIIaiDTT). Only at low concentrations of thromboplastin was FXIIaiDTT of FXI-deficient plasma significantly prolonged than that of normal plasma. Depletion of FXI from normal plasma prolonged its FXIIaiDTT and replenishment of FXI shortened it. FXIIaiDTTs of both FVIII-deficient and FIX-deficient plasma were remarkably prolonged, and addition of normal plasma dose-dependently shortened it. Furthermore, earlier alpha-thrombin inhibition was directly correlated with decreasing FXa generation. The amount of FXa production was: platelet-rich plasma > platelet-poor plasma > FXI-deficient plasma. Therefore, our findings from the FXIIaiDTT assays not only support the critical role of extrinsic pathway in blood coagulation initiation, but also demonstrate the importance of FXI as an amplifier of thrombin generation in thromboplastin-initiated coagulation.


Blood Coagulation Tests , Factor XI/physiology , Thromboplastin , Blood Coagulation/drug effects , Blood Platelets/metabolism , Blood Platelets/physiology , Factor VIII/pharmacology , Factor XI/pharmacology , Factor Xa/drug effects , Hemostatics , Humans , Partial Thromboplastin Time , Plant Proteins/pharmacology , Thrombin/pharmacology
16.
Curr Opin Hematol ; 7(5): 266-72, 2000 Sep.
Article En | MEDLINE | ID: mdl-10961575

Factor XI is a component of the intrinsic pathway of coagulation. A deficiency of factor XI is associated with a mild to moderate bleeding disorder especially from tissues with a high local fibrinolytic activity. In contrast, high levels of factor XI are a risk factor for venous thrombosis. The recent finding that factor XI can be activated by thrombin led to a revised model of coagulation. In this model the primary thrombin generation that results in fibrin formation takes place via the extrinsic pathway. Additional thrombin generation takes place inside the fibrin clot via the intrinsic pathway after the activation of factor XI by thrombin. High concentrations of thrombin are formed that are necessary for the activation of thrombin activatable fibrinolysis inhibitor (TAFI). Activated TAFI protects the fibrin clot against lysis. The role of factor XI in hemostasis can therefore be seen as a combination of procoagulant and antifibrinolytic actions. The new insights in the role of factor XI in coagulation and fibrinolysis may lead to new strategies for the treatment of thrombotic disorders.


Factor XI/physiology , Fibrinolysis/drug effects , Animals , Down-Regulation/drug effects , Factor XI/pharmacology , Humans , Thrombin/biosynthesis , Thrombin/metabolism , Venous Thrombosis/etiology
18.
Arterioscler Thromb Vasc Biol ; 19(1): 170-7, 1999 Jan.
Article En | MEDLINE | ID: mdl-9888880

Thrombin can activate factor XI in the presence of dextran sulfate or sulfatides. However, a physiological cofactor for thrombin activation of factor XI has not been identified. We examined this question in a cell-based, tissue factor-initiated model system. In the absence of factor XII, factor XI enhanced thrombin generation in this model. The effect on thrombin generation was reproduced by 2 to 5 pmol/L factor XIa. A specific inhibitor of factor XIIa did not diminish the effect of factor XI. Thus, factor XI can be activated in a model system that does not contain factor XIIa or nonphysiological cofactors. Preincubation of factor XI with activated platelets and thrombin or factor Xa enhanced subsequent thrombin generation in the model system. Preincubation of factor XI with thrombin or factor Xa, but without platelets, did not enhance thrombin generation, suggesting that these proteases might activate factor XI on platelet surfaces. Thrombin and factor Xa were then directly tested for their ability to activate factor XI. In the presence of dextran sulfate, thrombin or factor Xa activated factor XI. Thrombin, but not factor Xa, also cleaved detectable amounts of factor XI in the presence of activated platelets. Thus, thrombin activates enough factor XI to enhance subsequent thrombin generation in a model system. Platelet surfaces might provide the site for thrombin activation of functionally significant amounts of factor XI in vivo.


Blood Platelets/metabolism , Factor XII/physiology , Factor XI/metabolism , Platelet Activation , Thrombin/pharmacology , Anticoagulants/pharmacology , Antithrombin III/pharmacology , Factor IX/pharmacology , Factor V/pharmacology , Factor VIII/pharmacology , Factor X/pharmacology , Factor XI/pharmacology , Humans , Kininogens/pharmacology , Lipoproteins/pharmacology , Prothrombin/pharmacology , Thrombin/metabolism
19.
J Clin Invest ; 72(5): 1706-15, 1983 Nov.
Article En | MEDLINE | ID: mdl-6605369

Radioiodinated Factor IX was cleaved by a crude sonicate from leukocytes. In the absence of calcium, fragments of less than 15,000 mol wt were seen from reduced samples on gel electrophoresis. After digestion in 2 mM calcium, however, electrophoresis of reduced samples showed, in addition to low molecular weight fragments, protein bands corresponding in size to heavy and light chains of Factor XIa-activated Factor IX. The cleaving activity in leukocyte sonicates was inhibited by soybean trypsin inhibitor, but only to a small extent by aprotinin. Granulocyte elastase was isolated from purified polymorphonuclear leukocyte granules by affinity chromatography on soybean trypsin inhibitor-agarose and further chromatography on carboxymethyl cellulose. The purified fraction contained two isozymes on acidic gels which cleaved both an ester sensitive to elastase and radiolabeled Factor IX. These two activities were inhibited by elastase-specific chloromethyl ketone. The isolated protease fraction rapidly inactivated apparent Factor IX activity in a coagulant assay system. The degree of inactivation correlated with the amount of intact, radiolabeled Factor IX cleaved. As with the crude sonicate, generation of the larger heavy and light chain-sized fragments was dependent upon calcium. To assess directly the effect of elastase on Factor IX, an immunospecific, active site-directed assay was developed. In this assay, the sample was incubated with solid-phase antibody to Factor IX and the amount of activated product was detected as that which had complexed with radioiodinated antithrombin III. In this system, exposure of Factor IX to Factor XIa showed progressive increase in the ability to bind antithrombin III, whereas after elastase, Factor XIa was unable to generate antithrombin III binding. The elastase-degraded Factor IX did not inhibit activation of additional Factor IX in clotting assays. When Factor IXa was incubated with elastase, binding of antithrombin III was decreased, corresponding to appearance of low molecular weight fragments on parallel samples that were reduced and electrophoresed. These data are consistent with elastase inactivating Factor IX by cleaving bonds near, but distinct from, bonds cleaved by Factor XIa.


Factor IX/antagonists & inhibitors , Granulocytes/enzymology , Pancreatic Elastase/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antithrombin III/metabolism , Factor IX/metabolism , Factor IX/pharmacology , Factor IXa , Factor XI/pharmacology , Factor XIa , Humans , Isoenzymes/blood , Pancreatic Elastase/antagonists & inhibitors , Pancreatic Elastase/blood
20.
Thromb Res ; 29(4): 407-17, 1983 Feb 15.
Article En | MEDLINE | ID: mdl-6344314

In order to compare the relative potencies of plasma kallikrein, beta-Factor XIIa, Factor XIa and urokinase as plasminogen activators, plasminogen activation by these proteins was studied using a radiolabeled fibrin plate assay. Urokinase was approximately 20,000 times more active than kallikrein or Factor XIa and 300,000 times more active than beta-Factor XIIa. Kallikrein and Factor XIa were approximately equal in plasminogen activator activity and were 20 times more potent than beta-Factor XIIa.


Endopeptidases/pharmacology , Factor XII/pharmacology , Factor XI/pharmacology , Kallikreins/pharmacology , Peptide Fragments/pharmacology , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/pharmacology , Factor XIIa , Factor XIa , Fibrin/pharmacology , Humans
...