Semin Thromb Hemost
DOI: 10.1055/s-0043-57034
Review Article

Plasma Kallikrein as a Forgotten Clotting Factor

1   Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
,
2   Laboratory for Clinical Thrombosis and Haemostasis, Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
,
3   Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
,
4   Division of Hematology and UNC Blood Research Center, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
,
1   Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
› Author Affiliations
Funding H.P. acknowledges funding from the British Heart Foundation (SP/14/1/30717), the Wellcome Trust (110373), the Medical Research Council Confidence in Concept (MC_PC_14109), and Innovate UK (30084). J.E. acknowledges funding from the British Heart Foundation Program Grant no. RG/12/9/29775. N.S.K. acknowledges funding support from the National Institutes of Health (NIH), the National Heart, Lung, and Blood Institute (Grant # UO1HL117659).

Abstract

For decades, it was considered that plasma kallikrein's (PKa) sole function within the coagulation cascade is the activation of factor (F)XII. Until recently, the two key known activators of FIX within the coagulation cascade were activated FXI(a) and the tissue factor–FVII(a) complex. Simultaneously, and using independent experimental approaches, three groups identified a new branch of the coagulation cascade, whereby PKa can directly activate FIX. These key studies identified that (1) FIX or FIXa can bind with high affinity to either prekallikrein (PK) or PKa; (2) in human plasma, PKa can dose dependently trigger thrombin generation and clot formation independent of FXI; (3) in FXI knockout murine models treated with intrinsic pathway agonists, PKa activity results in increased formation of FIXa:AT complexes, indicating direct activation of FIX by PKa in vivo. These findings suggest that there is both a canonical (FXIa-dependent) and non-canonical (PKa-dependent) pathway of FIX activation. These three recent studies are described within this review, alongside historical data that hinted at the existence of this novel role of PKa as a coagulation clotting factor. The implications of direct PKa cleavage of FIX remain to be determined physiologically, pathophysiologically, and in the context of next-generation anticoagulants in development.



Publication History

Article published online:
18 April 2023

© 2023. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Osterud B, Laake K, Prydz H. The activation of human factor IX. Thromb Diath Haemorrh 1975; 33 (03) 553-563
  • 2 Osterud B, Bouma BN, Griffin JH. Human blood coagulation factor IX. Purification, properties, and mechanism of activation by activated factor XI. J Biol Chem 1978; 253 (17) 5946-5951
  • 3 Visser M, van Oerle R, Ten Cate H. et al. Plasma kallikrein contributes to coagulation in the absence of factor XI by activating factor IX. Arterioscler Thromb Vasc Biol 2020; 40 (01) 103-111
  • 4 Noubouossie DF, Henderson MW, Mooberry M. et al. Red blood cell microvesicles activate the contact system, leading to factor IX activation via 2 independent pathways. Blood 2020; 135 (10) 755-765
  • 5 Kearney KJ, Butler J, Posada OM. et al. Kallikrein directly interacts with and activates factor IX, resulting in thrombin generation and fibrin formation independent of Factor XI. Proc Natl Acad Sci U S A 2021; 118 (03) e2014810118
  • 6 MacFarlane RG. An enzyme cascade in the blood clotting mechanism, and its function as a biochemical amplifier. Nature 1964; 202: 498-499
  • 7 Davie EW, Ratnoff OD. Waterfall sequence for intrinsic blood clotting. Science 1964; 145 (3638): 1310-1312
  • 8 Shamanaev A, Litvak M, Gailani D. Recent advances in factor XII structure and function. Curr Opin Hematol 2022; 29 (05) 233-243
  • 9 Shamanaev A, Ivanov I, Sun MF. et al. Model for surface-dependent factor XII activation: the roles of factor XII heavy chain domains. Blood Adv 2022; 6 (10) 3142-3154
  • 10 Revak SD, Cochrane CG, Bouma BN, Griffin JH. Surface and fluid phase activities of two forms of activated Hageman factor produced during contact activation of plasma. J Exp Med 1978; 147 (03) 719-729
  • 11 Ivanov I, Matafonov A, Gailani D. Single-chain factor XII: a new form of activated factor XII. Curr Opin Hematol 2017; 24 (05) 411-418
  • 12 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
  • 13 Ivanov I, Verhamme IM, Sun MF. et al. Protease activity in single-chain prekallikrein. Blood 2020; 135 (08) 558-567
  • 14 Heestermans M, Naudin C, Mailer RK. et al. Identification of the factor XII contact activation site enables sensitive coagulation diagnostics. Nat Commun 2021; 12 (01) 5596
  • 15 Schmaier AH. The contact activation and kallikrein/kinin systems: pathophysiologic and physiologic activities. J Thromb Haemost 2016; 14 (01) 28-39
  • 16 Shariat-Madar Z, Mahdi F, Schmaier AH. Identification and characterization of prolylcarboxypeptidase as an endothelial cell prekallikrein activator. J Biol Chem 2002; 277 (20) 17962-17969
  • 17 Gittleman HR, Merkulova A, Alhalabi O. et al. A cross-sectional study of KLKB1 and PRCP polymorphisms in patient samples with cardiovascular disease. Front Med (Lausanne) 2016; 3: 17
  • 18 Joseph K, Tholanikunnel BG, Kaplan AP. Heat shock protein 90 catalyzes activation of the prekallikrein-kininogen complex in the absence of factor XII. Proc Natl Acad Sci U S A 2002; 99 (02) 896-900
  • 19 Singh PK, Chen ZL, Horn K, Norris EH. Blocking domain 6 of high molecular weight kininogen to understand intrinsic clotting mechanisms. Res Pract Thromb Haemost 2022; 6 (07) e12815
  • 20 Mohammed BM, Matafonov A, Ivanov I. et al. An update on factor XI structure and function. Thromb Res 2018; 161: 94-105
  • 21 Li C, Voos KM, Pathak M. et al. Plasma kallikrein structure reveals apple domain disc rotated conformation compared to factor XI. J Thromb Haemost 2019; 17 (05) 759-770
  • 22 Fujikawa K, Chung DW, Hendrickson LE, Davie EW. Amino acid sequence of human factor XI, a blood coagulation factor with four tandem repeats that are highly homologous with plasma prekallikrein. Biochemistry 1986; 25 (09) 2417-2424
  • 23 Ponczek MB, Shamanaev A, LaPlace A. et al. The evolution of factor XI and the kallikrein-kinin system. Blood Adv 2020; 4 (24) 6135-6147
  • 24 Sun Y, Gailani D. Identification of a factor IX binding site on the third apple domain of activated factor XI. J Biol Chem 1996; 271 (46) 29023-29028
  • 25 Geng Y, Verhamme IM, Messer A. et al. A sequential mechanism for exosite-mediated factor IX activation by factor XIa. J Biol Chem 2012; 287 (45) 38200-38209
  • 26 Gailani D, Geng Y, Verhamme I. et al. The mechanism underlying activation of factor IX by factor XIa. Thromb Res 2014; 133 (0 1, Suppl 1): S48-S51
  • 27 Enfield DL, Thompson AR. Cleavage and activation of human factor IX by serine proteases. Blood 1984; 64 (04) 821-831
  • 28 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 (07) 1341-1352
  • 29 Rosenthal RL, Dreskin OH, Rosenthal N. New hemophilia-like disease caused by deficiency of a third plasma thromboplastin factor. Proc Soc Exp Biol Med 1953; 82 (01) 171-174
  • 30 Duga S, Salomon O. Congenital factor XI deficiency: an update. Semin Thromb Hemost 2013; 39 (06) 621-631
  • 31 Santoro C, Di Mauro R, Baldacci E. et al. Bleeding phenotype and correlation with factor XI (FXI) activity in congenital FXI deficiency: results of a retrospective study from a single centre. Haemophilia 2015; 21 (04) 496-501
  • 32 Seligsohn U. Factor XI deficiency in humans. J Thromb Haemost 2009; 7 (Suppl. 01) 84-87
  • 33 Salloum-Asfar S, de la Morena-Barrio ME, Esteban J. et al. Assessment of two contact activation reagents for the diagnosis of congenital factor XI deficiency. Thromb Res 2018; 163: 64-70
  • 34 Owens III AP, Mackman N. Microparticles in hemostasis and thrombosis. Circ Res 2011; 108 (10) 1284-1297
  • 35 Rubin O, Delobel J, Prudent M. et al. Red blood cell-derived microparticles isolated from blood units initiate and propagate thrombin generation. Transfusion 2013; 53 (08) 1744-1754
  • 36 Gao Y, Lv L, Liu S, Ma G, Su Y. Elevated levels of thrombin-generating microparticles in stored red blood cells. Vox Sang 2013; 105 (01) 11-17
  • 37 Jy W, Johansen ME, Bidot Jr C, Horstman LL, Ahn YS. Red cell-derived microparticles (RMP) as haemostatic agent. Thromb Haemost 2013; 110 (04) 751-760
  • 38 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 (07) 1355-1362
  • 39 Prudent M, Crettaz D, Delobel J, Seghatchian J, Tissot JD, Lion N. Differences between calcium-stimulated and storage-induced erythrocyte-derived microvesicles. Transfus Apheresis Sci 2015; 53 (02) 153-158
  • 40 Satta N, Toti F, Feugeas O. et al. Monocyte vesiculation is a possible mechanism for dissemination of membrane-associated procoagulant activities and adhesion molecules after stimulation by lipopolysaccharide. J Immunol 1994; 153 (07) 3245-3255
  • 41 Mooberry MJ, Bradford R, Hobl EL, Lin FC, Jilma B, Key NS. Procoagulant microparticles promote coagulation in a factor XI-dependent manner in human endotoxemia. J Thromb Haemost 2016; 14 (05) 1031-1042
  • 42 van Beers EJ, Schaap MC, Berckmans RJ. et al; CURAMA Study Group. Circulating erythrocyte-derived microparticles are associated with coagulation activation in sickle cell disease. Haematologica 2009; 94 (11) 1513-1519
  • 43 Stief TW. Kallikrein activates prothrombin. Clin Appl Thromb Hemost 2008; 14 (01) 97-98
  • 44 Qian Y, Pan J, Zhou X, Weiser P, Lu H, Zhang L. Molecular mechanism underlines heparin-induced thrombocytopenia and thrombosis. Prog Mol Biol Transl Sci 2010; 93: 395-421
  • 45 Henderson MW, Lima F, Moraes CRP. et al. Contact and intrinsic coagulation pathways are activated and associated with adverse clinical outcomes in COVID-19. Blood Adv 2022; 6 (11) 3367-3377
  • 46 Sparkenbaugh EM, Henderson MW, Miller-Awe MD. et al. Factor XII contributes to thrombotic complications and vaso-occlusion in sickle cell disease. Blood 2023; blood.2022017074