Thromb Haemost 2010; 104(03): 514-522
DOI: 10.1160/TH09-10-0690
Blood Coagulation, Fibrinolysis and Cellular Haemostasis
Schattauer GmbH

In vitro effects of human neutrophil cathepsin G on thrombin generation: Both acceleration and decreased potential

Julien Perrin
1   Hématologie biologique, Pôle laboratoires, CHU Nancy, Vandœuvre-lès-Nancy, France
2   EA3452, Faculté de Pharmacie, Nancy Université, France
,
Thomas Lecompte
1   Hématologie biologique, Pôle laboratoires, CHU Nancy, Vandœuvre-lès-Nancy, France
3   Inserm U961, Nancy, France
,
Agnès Tournier
3   Inserm U961, Nancy, France
,
Lise Morlon
1   Hématologie biologique, Pôle laboratoires, CHU Nancy, Vandœuvre-lès-Nancy, France
,
Monique Marchand-Arvier
1   Hématologie biologique, Pôle laboratoires, CHU Nancy, Vandœuvre-lès-Nancy, France
,
Claude Vigneron
1   Hématologie biologique, Pôle laboratoires, CHU Nancy, Vandœuvre-lès-Nancy, France
2   EA3452, Faculté de Pharmacie, Nancy Université, France
› Author Affiliations
Further Information

Publication History

Received: 09 November 2009

Accepted after major revision: 11 May 2010

Publication Date:
23 November 2017 (online)

Summary

Cathepsin G (Cath G), a serine-protease found in neutrophils, has been reported to have effects that could either facilitate or impede coagulation. Thrombin generation (CAT method) was chosen to study its overall effect on the process, at a plasma concentration (240 nM) observed after neutrophil activation. Coagulation was triggered by tissue factor in the presence of platelets or phospholipid vesicles. To help identify potential targets of Cath G, plasma depleted of clotting factors or of inhibitors was used. Cath G induced a puzzling combination of two diverging effects of varying intensities depending on the phospholipid surface provided: accelerating the process under the three conditions (shortened clotting time by up to 30%), and impeding the process during the same thrombin generation time-course since thrombin peak and ETP (total thrombin potential) were decreased, up to 45% and 12%, respectively, suggestive of deficient prothrombinase. This is consistent with Cath G working on at least two targets in the coagulation cascade. Our data indicate that coagulation acceleration can be attributed neither to platelet activation and nor to activation of a clotting factor. When TFPI (tissue factor pathway inhibitor) was absent, no effect on lag time was observed and the anticoagulant activity of TFPI was decreased in the presence of Cath G. Consistent with the literature and the hypothesis of deficient prothrombinase, experiments using Russel’s Viper Venom indicate that the anticoagulant effect can be attributed to a deleterious effect on factor V. The clinical relevance of these findings deserves to be studied.

 
  • References

  • 1 Esmon CT. Inflammation and thrombosis. J Thromb Haemost 2003; 1: 1343-1348.
  • 2 Levi M. Cross-talk between coagulation and inflammation. Biomedical Progr 2003; 16: 8-14.
  • 3 Wagner DD. New links between inflammation and thrombosis. Arterioscler Thromb Vasc Biol 2005; 25: 1321-1324.
  • 4 Goldsack NR, Chambers RC, Dabbagh K. et al. Molecules in focus: Thrombin. Int J Biochem Cell Biol 1998; 30: 641-646.
  • 5 Krupiczojc MA, Scotton CJ, Chambers RC. Coagulation signalling following tissue injury: focus on the role of factor Xa. Int J Biochem Cell Biol 2008; 40: 1228-1237.
  • 6 Bouchard BA, Tracy PB. The participation of leukocytes in coagulant reactions. J Thromb Haemost 2003; 1: 464-469.
  • 7 Perrin J, Morlon L, Vigneron C. et al. Influence of polymorphonuclear leukocytes on the plasma clot formation as evaluated by thromboelastometry (ROTEM). Thromb Res 2008; 121: 647-652.
  • 8 Korkmaz B, Moreau T, Gauthier F. Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physiopathological functions. Biochimie 2008; 90: 227-242.
  • 9 Pham CT. Neutrophil serine proteases fine-tune the inflammatory response. Int J Biochem Cell Biol 2008; 40: 1317-1333.
  • 10 Chignard M, Selak MA, Smith JB. Direct evidence for the existence of a neutro-phil-derived platelet activator (neutrophilin). Proc Natl Acad Sci USA 1986; 83: 8609-8613.
  • 11 Selak MA, Chignard M, Smith JB. Cathepsin G is a strong platelet agonist released by neutrophils. Biochem J 1988; 251: 293-299.
  • 12 Renesto P, Chignard M. Neutrophil-mediated platelet activation: a key role for serine proteinases. Gen Pharmacol 1995; 26: 909-910.
  • 13 Cerletti C, Evangelista V, Molino M. et al. Platelet activation by polymorphonu-clear leukocytes: role of cathepsin G and P-selectin. Thromb Haemost 1995; 74: 218-223.
  • 14 Sambrano GR, Huang W, Faruqi T. et al. Cathepsin G activates protease-activated receptor-4 in human platelets. J Biol Chem 2000; 275: 6819-6823.
  • 15 Ossovskaya VS, Bunnett NW. Protease-activated receptors: contribution to physiology and disease. Physiol Rev 2004; 84: 579-621.
  • 16 Turkington PT. Cathepsin G a regulator of human vitamin K, dependent clotting factors and inhibitors. Thromb Res 1992; 67: 147-155.
  • 17 Brezniak DV, Brower MS, Witting JI. et al. Human alpha- to zeta-thrombin cleavage occurs with neutrophil cathepsin G or chymotrypsin while fibrinogen clotting activity is retained. Biochemistry 1990; 29: 3536-3542.
  • 18 Petersen LC, Bjørn SE, Nordfang O. Effect of leukocyte proteinases on tissue factor pathway inhibitor. Thromb Haemost 1992; 67: 537-541.
  • 19 Bagoly Z, Haramura G, Muszbek L. Down-regulation of activated factor XIII by polymorphonuclear granulocyte proteases within fibrin clot. Thromb Haemost 2007; 98: 359-367.
  • 20 Turkington PT. Cathepsin G activates human factor V in vitro. Thromb Res 1993; 72: 333-337.
  • 21 Allen DH, Tracy PB. Human coagulation factor V is activated to the functional cofactor by elastase and cathepsin G expressed at the monocyte surface. J Biol Chem 1995; 270: 1408-1415.
  • 22 Camire RM, Kalafatis M, Tracy PB. Proteolysis of factor V by cathepsin G and elastase indicates that cleavage at Arg1545 optimizes cofactor function by facilitating factor Xa binding. Biochemistry 1998; 37: 11896-11906.
  • 23 Gale AJ, Rozenshteyn D. Cathepsin G a leukocyte protease, activates coagulation factor VIII. Thromb Haemost 2008; 99: 44-51.
  • 24 Hemker HC. Recollections on thrombin generation. J Thromb Haemost 2008; 6: 219-226.
  • 25 Hemker HC, Giesen P, Al Dieri R. et al. Calibrated automated thrombin generation measurement in clotting plasma. Pathophysiol Haemost Thromb 2003; 33: 4-15.
  • 26 Renesto P, Chignard M. Enhancement of cathepsin G-induced platelet activation by leukocyte elastase: consequence for the neutrophil-mediated platelet activation. Blood 1993; 82: 139-144.
  • 27 Regnault V, Béguin S, Lecompte T. Calibrated automated thrombin generation in frozen-thawed platelet-rich plasma to detect hypercoagulability. Pathophysiol Haemost Thromb 2003; 33: 23-29.
  • 28 Hézard N, Bouaziz-Borgi L, Remy MG. et al. Utility of thrombin-generation assay in the screening of factor V G1691A (Leiden) and prothrombin G20210A mutations and protein S deficiency. Clin Chem 2006; 52: 665-670.
  • 29 Trossaërt M, Regnault V, Sigaud M. et al. Mild hemophilia A with factor VIII assay discrepancy: using thrombin generation assay to assess the bleeding phenotype. J Thromb Haemost 2008; 6: 486-493.
  • 30 MacFarlane RG. The coagulant action of Russell’s viper venom; the use of anti-venom in defining its reaction with a serum factor. Br J Haematol 1961; 7: 496-511.
  • 31 Regnault V, Hemker HC, Wahl D. et al. Phenotyping the haemostatic system by thrombography--potential for the estimation of thrombotic risk. Thromb Res 2004; 114: 539-545.
  • 32 Dargaud Y, Béguin S, Lienhart A. et al. Evaluation of thrombin generating capacity in plasma from patients with haemophilia A and B. Thromb Haemost 2005; 93: 475-480.
  • 33 van Hylckama Vlieg A, Christiansen SC, Luddington R. et al. Elevated endogenous thrombin potential is associated with an increased risk of a first deep venous thrombosis but not with the risk of recurrence. Br J Haematol 2007; 138: 769-774.
  • 34 Ghosh K, Mota L, Shetty S. et al. Spectrum of changes in endogenous thrombin potential due to heritable disorders of coagulation. Blood Coagul Fibrinolysis 2008; 19: 577-580.
  • 35 Al Dieri R, Peyvandi F, Santagostino E. et al. The thrombogram in rare inherited coagulation disorders: its relation to clinical bleeding. Thromb Haemost 2002; 88: 576-582.
  • 36 Duchemin J, Pan-Petesch B, Arnaud B. et al. Influence of coagulation factors and tissue factor concentration on the thrombin generation test in plasma. Thromb Haemost 2008; 99: 767-773.
  • 37 Dahm AE, Andersen TO, Rosendaal F. et al. A novel anticoagulant activity assay of tissue factor pathway inhibitor I (TFPI). J Thromb Haemost 2005; 3: 651-658.
  • 38 Fourrier F, Jourdain M, Tournois A. et al. Coagulation inhibitor substitution during sepsis. Intensive Care Med 1995; 21: S264-S268.
  • 39 Gando S, Kameue T, Matsuda N. et al. Serial changes in neutrophil-endothelial activation markers during the course of sepsis associated with disseminated intravascular coagulation. Thromb Res 2005; 116: 91-100.
  • 40 Falanga A, Marchetti M, Evangelista V. et al. Polymorphonuclear leukocyte activation and hemostasis in patients with essential thrombocythemia and polycythemia vera. Blood. 2000; 96: 4261-4266.
  • 41 Marchetti M, Falanga A. Leukocytosis, JAK2V617F mutation, and hemostasis in myeloproliferative disorders. Pathophysiol Haemost Thromb 2008; 36: 148-159.
  • 42 Xi M, Béguin S, Hemker HC. Importance of factor-IX-dependent prothrombinase formation--the Josso pathway--in clotting plasma. Haemostasis 1989; 19: 301-308.
  • 43 Hoffman M. A cell-based model of coagulation and the role of factor VIIa. Blood Rev 2003; 17: S1-S5.
  • 44 Mann KG. Thrombin formation. Chest 2003; 124: 4S-10S.
  • 45 Mann KG, Kalafatis M. Factor V: a combination of Dr Jekyll and Mr Hyde. Blood 2003; 101: 20-30.
  • 46 Steppich BA, Seitz I, Busch G. et al. Modulation of tissue factor and tissue factor pathway inhibitor-1 by neutrophil proteases. Thromb Haemost 2008; 100: 1068-1075.
  • 47 Collins PW, Macchiavello LI, Lewis SJ. et al. Global tests of haemostasis in critically ill patients with severe sepsis syndrome compared to controls. Br J Haematol 2006; 135: 220-227.
  • 48 Marchetti M, Castoldi E, Spronk HM. et al. Thrombin generation and activated protein C resistance in patients with essential thrombocythemia and polycythemia vera. Blood 2008; 112: 406-418.