Semin Thromb Hemost 2015; 41(06): 650-658
DOI: 10.1055/s-0035-1556730
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Coagulopathy of Acute Sepsis

Nicola Semeraro
1   Section of Experimental and Clinical Pathology, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
,
Concetta T. Ammollo
1   Section of Experimental and Clinical Pathology, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
,
Fabrizio Semeraro
1   Section of Experimental and Clinical Pathology, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
,
Mario Colucci
1   Section of Experimental and Clinical Pathology, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
› Author Affiliations
Further Information

Publication History

Publication Date:
25 August 2015 (online)

Abstract

Coagulopathy is common in acute sepsis and may range from subclinical activation of blood coagulation (hypercoagulability), which may contribute to venous thromboembolism, to acute disseminated intravascular coagulation, characterized by widespread microvascular thrombosis and consumption of platelets and coagulation proteins, eventually causing bleeding. The key event underlying this life-threatening complication is the overwhelming inflammatory host response to the pathogen leading to the overexpression of inflammatory mediators. The latter, along with the microorganism and its derivatives drive the major changes responsible for massive thrombin formation and fibrin deposition: (1) aberrant expression of tissue factor mainly by monocytes-macrophages, (2) impairment of anticoagulant pathways, orchestrated by dysfunctional endothelial cells (ECs), and (3) suppression of fibrinolysis because of the overproduction of plasminogen activator inhibitor-1 by ECs and thrombin-mediated activation of thrombin-activatable fibrinolysis inhibitor. Neutrophils and other cells, upon activation or death, release nuclear materials (neutrophil extracellular traps and/or their components such as histones, DNA, lysosomal enzymes, and High Mobility Group Box-1), which have toxic, proinflammatory and prothrombotic properties thus contributing to clotting dysregulation. The ensuing microvascular thrombosis–ischemia significantly contributes to tissue injury and multiple organ dysfunction syndromes. These insights into the pathogenesis of sepsis-associated coagulopathy may have implications for the development of new diagnostic and therapeutic tools.

 
  • References

  • 1 Levi M, Schultz M, van der Poll T. Disseminated intravascular coagulation in infectious disease. Semin Thromb Hemost 2010; 36 (4) 367-377
  • 2 Semeraro N, Ammollo CT, Semeraro F, Colucci M. Sepsis-associated disseminated intravascular coagulation and thromboembolic disease. Mediterr J Hematol Infect Dis 2010; 2 (3) e2010024
  • 3 Smeeth L, Cook C, Thomas S, Hall AJ, Hubbard R, Vallance P. Risk of deep vein thrombosis and pulmonary embolism after acute infection in a community setting. Lancet 2006; 367 (9516) 1075-1079
  • 4 Alikhan R, Spyropoulos AC. Epidemiology of venous thromboembolism in cardiorespiratory and infectious disease. Am J Med 2008; 121 (11) 935-942
  • 5 Wang H, Ma S. The cytokine storm and factors determining the sequence and severity of organ dysfunction in multiple organ dysfunction syndrome. Am J Emerg Med 2008; 26 (6) 711-715
  • 6 Levi M, Schultz M, van der Poll T. Sepsis and thrombosis. Semin Thromb Hemost 2013; 39 (5) 559-566
  • 7 Levi M, van der Poll T, Schultz M. Infection and inflammation as risk factors for thrombosis and atherosclerosis. Semin Thromb Hemost 2012; 38 (5) 506-514
  • 8 Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 2008; 8 (10) 776-787
  • 9 Monroe DM, Key NS. The tissue factor-factor VIIa complex: procoagulant activity, regulation, and multitasking. J Thromb Haemost 2007; 5 (6) 1097-1105
  • 10 Rezaie AR. Protease-activated receptor signalling by coagulation proteases in endothelial cells. Thromb Haemost 2014; 112 (5) 876-882
  • 11 Xu J, Zhang X, Pelayo R , et al. Extracellular histones are major mediators of death in sepsis. Nat Med 2009; 15 (11) 1318-1321
  • 12 Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood 2014; 123 (18) 2768-2776
  • 13 Pawlinski R, Mackman N. Cellular sources of tissue factor in endotoxemia and sepsis. Thromb Res 2010; 125 (Suppl. 01) S70-S73
  • 14 Gando S, Nanzaki S, Sasaki S, Kemmotsu O. Significant correlations between tissue factor and thrombin markers in trauma and septic patients with disseminated intravascular coagulation. Thromb Haemost 1998; 79 (6) 1111-1115
  • 15 Østerud B, Bjørklid E. Sources of tissue factor. Semin Thromb Hemost 2006; 32 (1) 11-23
  • 16 Maugeri N, Brambilla M, Camera M , et al. Human polymorphonuclear leukocytes produce and express functional tissue factor upon stimulation. J Thromb Haemost 2006; 4 (6) 1323-1330
  • 17 Camera M, Frigerio M, Toschi V , et al. Platelet activation induces cell-surface immunoreactive tissue factor expression, which is modulated differently by antiplatelet drugs. Arterioscler Thromb Vasc Biol 2003; 23 (9) 1690-1696
  • 18 Panes O, Matus V, Sáez CG, Quiroga T, Pereira J, Mezzano D. Human platelets synthesize and express functional tissue factor. Blood 2007; 109 (12) 5242-5250
  • 19 Østerud B. Tissue factor expression in blood cells. Thromb Res 2010; 125 (Suppl. 01) S31-S34
  • 20 Semeraro N, Colucci M. Tissue factor in health and disease. Thromb Haemost 1997; 78 (1) 759-764
  • 21 Pawlinski R, Wang JG, Owens III AP , et al. Hematopoietic and nonhematopoietic cell tissue factor activates the coagulation cascade in endotoxemic mice. Blood 2010; 116 (5) 806-814
  • 22 Franco RF, de Jonge E, Dekkers PE , et al. The in vivo kinetics of tissue factor messenger RNA expression during human endotoxemia: relationship with activation of coagulation. Blood 2000; 96 (2) 554-559
  • 23 Aras O, Shet A, Bach RR , et al. Induction of microparticle- and cell-associated intravascular tissue factor in human endotoxemia. Blood 2004; 103 (12) 4545-4553
  • 24 Wang JG, Manly D, Kirchhofer D, Pawlinski R, Mackman N. Levels of microparticle tissue factor activity correlate with coagulation activation in endotoxemic mice. J Thromb Haemost 2009; 7 (7) 1092-1098
  • 25 van Hinsbergh VW. Endothelium—role in regulation of coagulation and inflammation. Semin Immunopathol 2012; 34 (1) 93-106
  • 26 Collier ME, Mah PM, Xiao Y, Maraveyas A, Ettelaie C. Microparticle-associated tissue factor is recycled by endothelial cells resulting in enhanced surface tissue factor activity. Thromb Haemost 2013; 110 (5) 966-976
  • 27 Rangarajan S, Kessler C, Aledort L. The clinical implications of ADAMTS13 function: the perspectives of haemostaseologists. Thromb Res 2013; 132 (4) 403-407
  • 28 Claus RA, Bockmeyer CL, Budde U , et al. Variations in the ratio between von Willebrand factor and its cleaving protease during systemic inflammation and association with severity and prognosis of organ failure. Thromb Haemost 2009; 101 (2) 239-247
  • 29 Rondina MT, Schwertz H, Harris ES , et al. The septic milieu triggers expression of spliced tissue factor mRNA in human platelets. J Thromb Haemost 2011; 9 (4) 748-758
  • 30 Björkqvist J, Nickel KF, Stavrou E, Renné T. In vivo activation and functions of the protease factor XII. Thromb Haemost 2014; 112 (5) 868-875
  • 31 van der Poll T, de Jonge E, Levi M. Regulatory role of cytokines in disseminated intravascular coagulation. Semin Thromb Hemost 2001; 27 (6) 639-651
  • 32 Silasi-Mansat R, Zhu H, Popescu NI , et al. Complement inhibition decreases the procoagulant response and confers organ protection in a baboon model of Escherichia coli sepsis. Blood 2010; 116 (6) 1002-1010
  • 33 Semeraro N, Colucci M. Endothelial cell perturbation and disseminated intravascular coagulation. In: ten Cate H, Levi M, , eds. Molecular Mechanisms of Disseminated Intravascular Coagulation. Georgetown: Landes Bioscience; 2003: 156-190
  • 34 Esmon CT. Inflammation and thrombosis. J Thromb Haemost 2003; 1 (7) 1343-1348
  • 35 Faust SN, Levin M, Harrison OB , et al. Dysfunction of endothelial protein C activation in severe meningococcal sepsis. N Engl J Med 2001; 345 (6) 408-416
  • 36 Macias WL, Nelson DR. Severe protein C deficiency predicts early death in severe sepsis. Crit Care Med 2004; 32 (5, Suppl): S223-S228
  • 37 Liaw PC, Esmon CT, Kahnamoui K , et al. Patients with severe sepsis vary markedly in their ability to generate activated protein C. Blood 2004; 104 (13) 3958-3964
  • 38 Mosnier LO, Zlokovic BV, Griffin JH. The cytoprotective protein C pathway. Blood 2007; 109 (8) 3161-3172
  • 39 Tang H, Ivanciu L, Popescu N , et al. Sepsis-induced coagulation in the baboon lung is associated with decreased tissue factor pathway inhibitor. Am J Pathol 2007; 171 (3) 1066-1077
  • 40 Gando S. Role of fibrinolysis in sepsis. Semin Thromb Hemost 2013; 39 (4) 392-399
  • 41 Semeraro N, Ammollo CT, Semeraro F, Colucci M. Sepsis, thrombosis and organ dysfunction. Thromb Res 2012; 129 (3) 290-295
  • 42 Wolberg AS. Thrombin generation and fibrin clot structure. Blood Rev 2007; 21 (3) 131-142
  • 43 Mosnier LO, Bouma BN. Regulation of fibrinolysis by thrombin activatable fibrinolysis inhibitor, an unstable carboxypeptidase B that unites the pathways of coagulation and fibrinolysis. Arterioscler Thromb Vasc Biol 2006; 26 (11) 2445-2453
  • 44 Campbell RA, Overmyer KA, Selzman CH, Sheridan BC, Wolberg AS. Contributions of extravascular and intravascular cells to fibrin network formation, structure, and stability. Blood 2009; 114 (23) 4886-4896
  • 45 Mutch NJ, Engel R, Uitte de Willige S, Philippou H, Ariëns RA. Polyphosphate modifies the fibrin network and down-regulates fibrinolysis by attenuating binding of tPA and plasminogen to fibrin. Blood 2010; 115 (19) 3980-3988
  • 46 Carrieri C, Galasso R, Semeraro F, Ammollo CT, Semeraro N, Colucci M. The role of thrombin activatable fibrinolysis inhibitor and factor XI in platelet-mediated fibrinolysis resistance: a thromboelastographic study in whole blood. J Thromb Haemost 2011; 9 (1) 154-162
  • 47 Semeraro F, Ammollo CT, Semeraro N, Colucci M. Tissue factor-expressing monocytes inhibit fibrinolysis through a TAFI-mediated mechanism, and make clots resistant to heparins. Haematologica 2009; 94 (6) 819-826
  • 48 Emonts M, de Bruijne EL, Guimarães AH , et al. Thrombin-activatable fibrinolysis inhibitor is associated with severity and outcome of severe meningococcal infection in children. J Thromb Haemost 2008; 6 (2) 268-276
  • 49 Brinkmann V, Zychlinsky A. Neutrophil extracellular traps: is immunity the second function of chromatin?. J Cell Biol 2012; 198 (5) 773-783
  • 50 Clark SR, Ma AC, Tavener SA , et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 2007; 13 (4) 463-469
  • 51 Jahr S, Hentze H, Englisch S , et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001; 61 (4) 1659-1665
  • 52 Lapponi MJ, Carestia A, Landoni VI , et al. Regulation of neutrophil extracellular trap formation by anti-inflammatory drugs. J Pharmacol Exp Ther 2013; 345 (3) 430-437
  • 53 Fuchs TA, Brill A, Duerschmied D , et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107 (36) 15880-15885
  • 54 Kambas K, Mitroulis I, Apostolidou E , et al. Autophagy mediates the delivery of thrombogenic tissue factor to neutrophil extracellular traps in human sepsis. PLoS ONE 2012; 7 (9) e45427
  • 55 Carestia A, Rivadeneyra L, Romaniuk MA, Fondevila C, Negrotto S, Schattner M. Functional responses and molecular mechanisms involved in histone-mediated platelet activation. Thromb Haemost 2013; 110 (5) 1035-1045
  • 56 Semeraro F, Ammollo CT, Esmon NL, Esmon CT. Histones induce phosphatidylserine exposure and a procoagulant phenotype in human red blood cells. J Thromb Haemost 2014; 12 (10) 1697-1702
  • 57 Semeraro F, Ammollo CT, Morrissey JH , et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood 2011; 118 (7) 1952-1961
  • 58 Ammollo CT, Semeraro F, Xu J, Esmon NL, Esmon CT. Extracellular histones increase plasma thrombin generation by impairing thrombomodulin-dependent protein C activation. J Thromb Haemost 2011; 9 (9) 1795-1803
  • 59 Wildhagen KC, García de Frutos P, Reutelingsperger CP , et al. Nonanticoagulant heparin prevents histone-mediated cytotoxicity in vitro and improves survival in sepsis. Blood 2014; 123 (7) 1098-1101
  • 60 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; 104 (15) 6388-6393
  • 61 Faraday N, Schunke K, Saleem S , et al. Cathepsin G-dependent modulation of platelet thrombus formation in vivo by blood neutrophils. PLoS ONE 2013; 8 (8) e71447
  • 62 Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 2013; 13 (1) 34-45
  • 63 Arai Y, Yamashita K, Mizugishi K , et al. Serum neutrophil extracellular trap levels predict thrombotic microangiopathy after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2013; 19 (12) 1683-1689
  • 64 Sunden-Cullberg J, Norrby-Teglund A, Treutiger CJ. The role of high mobility group box-1 protein in severe sepsis. Curr Opin Infect Dis 2006; 19 (3) 231-236
  • 65 Ito T, Kawahara K, Nakamura T , et al. High-mobility group box 1 protein promotes development of microvascular thrombosis in rats. J Thromb Haemost 2007; 5 (1) 109-116
  • 66 Lv B, Wang H, Tang Y, Fan Z, Xiao X, Chen F. High-mobility group box 1 protein induces tissue factor expression in vascular endothelial cells via activation of NF-kappaB and Egr-1. Thromb Haemost 2009; 102 (2) 352-359
  • 67 Maugeri N, Campana L, Gavina M , et al. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J Thromb Haemost 2014; 12 (12) 2074-2088
  • 68 Hotchkiss RS, Nicholson DW. Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol 2006; 6 (11) 813-822
  • 69 Zeerleder S, Zwart B, Wuillemin WA , et al. Elevated nucleosome levels in systemic inflammation and sepsis. Crit Care Med 2003; 31 (7) 1947-1951
  • 70 Ekaney ML, Otto GP, Sossdorf M , et al. Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation. Crit Care 2014; 18 (5) 543
  • 71 Rhodes A, Wort SJ, Thomas H, Collinson P, Bennett ED. Plasma DNA concentration as a predictor of mortality and sepsis in critically ill patients. Crit Care 2006; 10 (2) R60
  • 72 Levi M, Meijers JC. DIC: which laboratory tests are most useful. Blood Rev 2011; 25 (1) 33-37
  • 73 Page AV, Liles WC. Biomarkers of endothelial activation/dysfunction in infectious diseases. Virulence 2013; 4 (6) 507-516
  • 74 Chen Q, Ye L, Jin Y , et al. Circulating nucleosomes as a predictor of sepsis and organ dysfunction in critically ill patients. Int J Infect Dis 2012; 16 (7) e558-e564
  • 75 Hatada T, Wada H, Nobori T , et al. Plasma concentrations and importance of High Mobility Group Box protein in the prognosis of organ failure in patients with disseminated intravascular coagulation. Thromb Haemost 2005; 94 (5) 975-979
  • 76 Ranieri VM, Thompson BT, Barie PS , et al; PROWESS-SHOCK Study Group. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med 2012; 366 (22) 2055-2064
  • 77 Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ , et al. Sepsis: multiple abnormalities, heterogeneous responses, and evolving understanding. Physiol Rev 2013; 93 (3) 1247-1288
  • 78 Iba T, Gando S, Thachil J. Anticoagulant therapy for sepsis-associated disseminated intravascular coagulation: the view from Japan. J Thromb Haemost 2014; 12 (7) 1010-1019
  • 79 Nakahara M, Ito T, Kawahara K , et al. Recombinant thrombomodulin protects mice against histone-induced lethal thromboembolism. PLoS ONE 2013; 8 (9) e75961
  • 80 Ito T, Kawahara K, Okamoto K , et al. Proteolytic cleavage of high mobility group box 1 protein by thrombin-thrombomodulin complexes. Arterioscler Thromb Vasc Biol 2008; 28 (10) 1825-1830
  • 81 Vlahos R, Stambas J, Bozinovski S, Broughton BR, Drummond GR, Selemidis S. Inhibition of Nox2 oxidase activity ameliorates influenza A virus-induced lung inflammation. PLoS Pathog 2011; 7 (2) e1001271
  • 82 Jiménez-Alcázar M, Napirei M, Panda R , et al. Impaired DNase1-mediated degradation of neutrophil extracellular traps is associated with acute thrombotic microangiopathies. J Thromb Haemost 2015; 13 (5) 732-742