Semin Thromb Hemost
DOI: 10.1055/s-0043-1768936
Review Article

Inhibitors of Polyphosphate and Neutrophil Extracellular Traps

Sreeparna Vappala
1   Department of Pathology and Laboratory Medicine, Centre for Blood Research, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
,
Stephanie A. Smith
2   Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
,
Jayachandran N. Kizhakkedathu
1   Department of Pathology and Laboratory Medicine, Centre for Blood Research, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
3   Department of Chemistry, School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
,
James H. Morrissey
2   Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
› Author Affiliations
Funding This work was supported by National Institutes of Health grant R35 HL135823 (J.H.M.), Canadian Institutes of Health Research grant 201610PJT (J.N.K.), and Natural Sciences and Engineering Council of Canada grant RGPIN-2018-03828 (J.N.K.). J.N.K. is a Tier 1 Canada Research Chair in Immunomodulating Materials and Immunotherapy.

Abstract

The contact pathway of blood clotting has received intense interest in recent years as studies have linked it to thrombosis, inflammation, and innate immunity. Because the contact pathway plays little to no role in normal hemostasis, it has emerged as a potential target for safer thromboprotection, relative to currently approved antithrombotic drugs which all target the final common pathway of blood clotting. Research since the mid-2000s has identified polyphosphate, DNA, and RNA as important triggers of the contact pathway with roles in thrombosis, although these molecules also modulate blood clotting and inflammation via mechanisms other than the contact pathway of the clotting cascade. The most significant source of extracellular DNA in many disease settings is in the form of neutrophil extracellular traps (NETs), which have been shown to contribute to incidence and severity of thrombosis. This review summarizes known roles of extracellular polyphosphate and nucleic acids in thrombosis, with an emphasis on novel agents under current development that target the prothrombotic activities of polyphosphate and NETs.



Publication History

Article published online:
16 May 2023

© 2023. Thieme. All rights reserved.

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

 
  • References

  • 1 Juang LJ, Mazinani N, Novakowski SK. et al. Coagulation factor XII contributes to hemostasis when activated by soil in wounds. Blood Adv 2020; 4 (08) 1737-1745
  • 2 Rao NN, Gómez-García MR, Kornberg A. Inorganic polyphosphate: essential for growth and survival. Annu Rev Biochem 2009; 78: 605-647
  • 3 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; 103 (04) 903-908
  • 4 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
  • 5 Gansler J, Jaax M, Leiting S. et al. Structural requirements for the procoagulant activity of nucleic acids. PLoS One 2012; 7 (11) e50399
  • 6 Bhagirath VC, Dwivedi DJ, Liaw PC. Comparison of the proinflammatory and procoagulant properties of nuclear, mitochondrial, and bacterial DNA. Shock 2015; 44 (03) 265-271
  • 7 Gould TJ, Vu TT, Stafford AR. et al. Cell-free DNA modulates clot structure and impairs fibrinolysis in sepsis. Arterioscler Thromb Vasc Biol 2015; 35 (12) 2544-2553
  • 8 Fuchs T, Brill A, Dürschmied D. et al. Neutrophil extracellular traps induce platelet adhesion and thrombus formation. Blood 2009; 114: 1345
  • 9 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
  • 10 Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 2012; 32 (08) 1777-1783
  • 11 Martinod K, Demers M, Fuchs TA. et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A 2013; 110 (21) 8674-8679
  • 12 Morrissey JH, Smith SA. Polyphosphate as modulator of hemostasis, thrombosis, and inflammation. J Thromb Haemost 2015; 13 (0 1, Suppl 1): S92-S97
  • 13 Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood 2014; 123 (18) 2768-2776
  • 14 Demers M, Wagner DD. NETosis: a new factor in tumor progression and cancer-associated thrombosis. Semin Thromb Hemost 2014; 40 (03) 277-283
  • 15 Long AT, Kenne E, Jung R, Fuchs TA, Renné T. Contact system revisited: an interface between inflammation, coagulation, and innate immunity. J Thromb Haemost 2016; 14 (03) 427-437
  • 16 Müller F, Gailani D, Renné T. Factor XI and XII as antithrombotic targets. Curr Opin Hematol 2011; 18 (05) 349-355
  • 17 Kokoye Y, Ivanov I, Cheng Q. et al. A comparison of the effects of factor XII deficiency and prekallikrein deficiency on thrombus formation. Thromb Res 2016; 140: 118-124
  • 18 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; 105 (02) 269-273
  • 19 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; 111 (08) 4113-4117
  • 20 He R, Chen D, He S. Factor XI: hemostasis, thrombosis, and antithrombosis. Thromb Res 2012; 129 (05) 541-550
  • 21 Key NS. Epidemiologic and clinical data linking factors XI and XII to thrombosis. Hematology (Am Soc Hematol Educ Program) 2014; 2014 (01) 66-70
  • 22 Büller HR, Bethune C, Bhanot S. et al; FXI-ASO TKA Investigators. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N Engl J Med 2015; 372 (03) 232-240
  • 23 Verhamme P, Yi BA, Segers A. et al; ANT-005 TKA Investigators. Abelacimab for prevention of venous thromboembolism. N Engl J Med 2021; 385 (07) 609-617
  • 24 Labberton L, Kenne E, Renné T. New agents for thromboprotection. A role for factor XII and XIIa inhibition. Hamostaseologie 2015; 35 (04) 338-350
  • 25 Matafonov A, Leung PY, Gailani AE. et al. Factor XII inhibition reduces thrombus formation in a primate thrombosis model. Blood 2014; 123 (11) 1739-1746
  • 26 Yamagata Y, Watanabe H, Saitoh M, Namba T. Volcanic production of polyphosphates and its relevance to prebiotic evolution. Nature 1991; 352 (6335): 516-519
  • 27 Kornberg A, Rao NN, Ault-Riché D. Inorganic polyphosphate: a molecule of many functions. Annu Rev Biochem 1999; 68: 89-125
  • 28 Moreno SN, Docampo R. Polyphosphate and its diverse functions in host cells and pathogens. PLoS Pathog 2013; 9 (05) e1003230
  • 29 Keasling JD. Regulation of intracellular toxic metals and other cations by hydrolysis of polyphosphate. Ann N Y Acad Sci 1997; 829: 242-249
  • 30 Gray MJ, Wholey WY, Wagner NO. et al. Polyphosphate is a primordial chaperone. Mol Cell 2014; 53 (05) 689-699
  • 31 Kumble KD, Kornberg A. Inorganic polyphosphate in mammalian cells and tissues. J Biol Chem 1995; 270 (11) 5818-5822
  • 32 Leyhausen G, Lorenz B, Zhu H. et al. Inorganic polyphosphate in human osteoblast-like cells. J Bone Miner Res 1998; 13 (05) 803-812
  • 33 Moreno-Sanchez D, Hernandez-Ruiz L, Ruiz FA, Docampo R. Polyphosphate is a novel pro-inflammatory regulator of mast cells and is located in acidocalcisomes. J Biol Chem 2012; 287 (34) 28435-28444
  • 34 Nickel KF, Ronquist G, Langer F. et al. The polyphosphate-factor XII pathway drives coagulation in prostate cancer-associated thrombosis. Blood 2015; 126 (11) 1379-1389
  • 35 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; 279 (43) 44250-44257
  • 36 Müller F, Mutch NJ, Schenk WA. et al. Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo. Cell 2009; 139 (06) 1143-1156
  • 37 Bae JS, Lee W, Rezaie AR. Polyphosphate elicits pro-inflammatory responses that are counteracted by activated protein C in both cellular and animal models. J Thromb Haemost 2012; 10 (06) 1145-1151
  • 38 Dinarvand P, Hassanian SM, Qureshi SH. et al. Polyphosphate amplifies proinflammatory responses of nuclear proteins through interaction with receptor for advanced glycation end products and P2Y1 purinergic receptor. Blood 2014; 123 (06) 935-945
  • 39 Choi SH, Smith SA, Morrissey JH. Polyphosphate accelerates factor V activation by factor XIa. Thromb Haemost 2015; 113 (03) 599-604
  • 40 Smith SA, Morrissey JH. Polyphosphate enhances fibrin clot structure. Blood 2008; 112 (07) 2810-2816
  • 41 Choi SH, Smith SA, Morrissey JH. Polyphosphate is a cofactor for the activation of factor XI by thrombin. Blood 2011; 118 (26) 6963-6970
  • 42 Smith SA, Choi SH, Davis-Harrison R. et al. Polyphosphate exerts differential effects on blood clotting, depending on polymer size. Blood 2010; 116 (20) 4353-4359
  • 43 Arredondo C, Cefaliello C, Dyrda A. et al. Excessive release of inorganic polyphosphate by ALS/FTD astrocytes causes non-cell-autonomous toxicity to motoneurons. Neuron 2022; 110 (10) 1656-1670.e12
  • 44 Stotz SC, Scott LO, Drummond-Main C. et al. Inorganic polyphosphate regulates neuronal excitability through modulation of voltage-gated channels. Mol Brain 2014; 7: 42
  • 45 Holmström KM, Marina N, Baev AY, Wood NW, Gourine AV, Abramov AY. Signalling properties of inorganic polyphosphate in the mammalian brain. Nat Commun 2013; 4: 1362
  • 46 Smith SA, Morrissey JH. 2013 scientific sessions Sol Sherry distinguished lecture in thrombosis: polyphosphate: a novel modulator of hemostasis and thrombosis. Arterioscler Thromb Vasc Biol 2015; 35 (06) 1298-1305
  • 47 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
  • 48 Baker CJ, Smith SA, Morrissey JH. Polyphosphate in thrombosis, hemostasis, and inflammation. Res Pract Thromb Haemost 2018; 3 (01) 18-25
  • 49 Bolesch DG, Keasling JD. Polyphosphate binding and chain length recognition of Escherichia coli exopolyphosphatase. J Biol Chem 2000; 275 (43) 33814-33819
  • 50 Saito K, Ohtomo R, Kuga-Uetake Y, Aono T, Saito M. Direct labeling of polyphosphate at the ultrastructural level in Saccharomyces cerevisiae by using the affinity of the polyphosphate binding domain of Escherichia coli exopolyphosphatase. Appl Environ Microbiol 2005; 71 (10) 5692-5701
  • 51 Werner TP, Amrhein N, Freimoser FM. Specific localization of inorganic polyphosphate (poly P) in fungal cell walls by selective extraction and immunohistochemistry. Fungal Genet Biol 2007; 44 (09) 845-852
  • 52 Choi SH, Smith SA, Morrissey JH. Platelet polyphosphate enhances factor XI activation by thrombin. Blood 2011; 118: 174-175
  • 53 Smith SA, Choi SH, Collins JN, Travers RJ, Cooley BC, Morrissey JH. Inhibition of polyphosphate as a novel strategy for preventing thrombosis and inflammation. Blood 2012; 120 (26) 5103-5110
  • 54 Labberton L, Kenne E, Long AT. et al. Neutralizing blood-borne polyphosphate in vivo provides safe thromboprotection. Nat Commun 2016; 7: 12616
  • 55 Jain K, Kesharwani P, Gupta U, Jain NK. Dendrimer toxicity: let's meet the challenge. Int J Pharm 2010; 394 (1-2): 122-142
  • 56 Jones CF, Campbell RA, Brooks AE. et al. Cationic PAMAM dendrimers aggressively initiate blood clot formation. ACS Nano 2012; 6 (11) 9900-9910
  • 57 Malik N, Wiwattanapatapee R, Klopsch R. et al. Dendrimers: relationship between structure and biocompatibility in vitro, and preliminary studies on the biodistribution of 125I-labelled polyamidoamine dendrimers in vivo. J Control Release 2000; 65 (1-2): 133-148
  • 58 Shenoi RA, Kalathottukaren MT, Travers RJ. et al. Affinity-based design of a synthetic universal reversal agent for heparin anticoagulants. Sci Transl Med 2014; 6 (260) 260ra150
  • 59 Abbina S, La CC, Vappala S. et al. Influence of steric shield on biocompatibility and antithrombotic activity of dendritic polyphosphate inhibitor. Mol Pharm 2022; 19 (06) 1853-1865
  • 60 Travers RJ, Shenoi RA, Kalathottukaren MT, Kizhakkedathu JN, Morrissey JH. Nontoxic polyphosphate inhibitors reduce thrombosis while sparing hemostasis. Blood 2014; 124 (22) 3183-3190
  • 61 Ni Ainle F, Preston RJS, Jenkins PV. et al. Protamine sulfate down-regulates thrombin generation by inhibiting factor V activation. Blood 2009; 114 (08) 1658-1665
  • 62 La CC, Smith SA, Vappala S. et al. Smart thrombosis inhibitors without bleeding side effects via charge tunable ligand design. Nat Commun 2023; 14: 2177
  • 63 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977; 37 (03) 646-650
  • 64 Singh N, Gupta S, Pandey RM, Chauhan SS, Saraya A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Invest 2015; 33 (03) 78-85
  • 65 Wen F, Shen A, Choi A, Gerner EW, Shi J. Extracellular DNA in pancreatic cancer promotes cell invasion and metastasis. Cancer Res 2013; 73 (14) 4256-4266
  • 66 Todorova VK, Hsu PC, Wei JY. et al. Biomarkers of inflammation, hypercoagulability and endothelial injury predict early asymptomatic doxorubicin-induced cardiotoxicity in breast cancer patients. Am J Cancer Res 2020; 10 (09) 2933-2945
  • 67 Swystun LL, Mukherjee S, Liaw PC. Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus. J Thromb Haemost 2011; 9 (11) 2313-2321
  • 68 Goswami J, MacArthur TA, Mahony C. et al. DNase-mediated dissolution of neutrophil extracellular traps accelerates in vitro thrombin generation kinetics in trauma patients. Shock 2022; 58 (03) 217-223
  • 69 Rainer TH, Wong LK, Lam W. et al. Prognostic use of circulating plasma nucleic acid concentrations in patients with acute stroke. Clin Chem 2003; 49 (04) 562-569
  • 70 Lo YM, Rainer TH, Chan LY, Hjelm NM, Cocks RA. Plasma DNA as a prognostic marker in trauma patients. Clin Chem 2000; 46 (03) 319-323
  • 71 Diaz JA, Fuchs TA, Jackson TO. et al; For the Michigan Research Venous Group*. Plasma DNA is elevated in patients with deep vein thrombosis. J Vasc Surg Venous Lymphat Disord 2013; 1 (04) 341-348.e1
  • 72 Smith SA, Baker CJ, Gajsiewicz JM, Morrissey JH. Silica particles contribute to the procoagulant activity of DNA and polyphosphate isolated using commercial kits. Blood 2017; 130 (01) 88-91
  • 73 Smith SA, Gajsiewicz JM, Morrissey JH. Ability of polyphosphate and nucleic acids to trigger blood clotting: some observations and caveats. Front Med (Lausanne) 2018; 5: 107
  • 74 Beckmann L, Voigtlaender M, Rolling CC, Schulenkorf A, Bokemeyer C, Langer F. Myeloperoxidase has no effect on the low procoagulant activity of silica-free DNA. Thromb Res 2021; 203: 36-45
  • 75 Medeiros SK, Zafar N, Liaw PC, Kim PY. Purification of silica-free DNA and characterization of its role in coagulation. J Thromb Haemost 2019; 17 (11) 1860-1865
  • 76 Oehmcke S, Mörgelin M, Herwald H. Activation of the human contact system on neutrophil extracellular traps. J Innate Immun 2009; 1 (03) 225-230
  • 77 Xu J, Zhang X, Pelayo R. et al. Extracellular histones are major mediators of death in sepsis. Nat Med 2009; 15 (11) 1318-1321
  • 78 Xu J, Zhang X, Monestier M, Esmon NL, Esmon CT. Extracellular histones are mediators of death through TLR2 and TLR4 in mouse fatal liver injury. J Immunol 2011; 187 (05) 2626-2631
  • 79 Gould TJ, Vu TT, Swystun LL. et al. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler Thromb Vasc Biol 2014; 34 (09) 1977-1984
  • 80 Brill A, Fuchs TA, Savchenko AS. et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 2012; 10 (01) 136-144
  • 81 Varjú I, Longstaff C, Szabó L. et al. DNA, histones and neutrophil extracellular traps exert anti-fibrinolytic effects in a plasma environment. Thromb Haemost 2015; 113 (06) 1289-1298
  • 82 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 (09) 1795-1803
  • 83 Saravanan R, Choong YK, Lim CH, Lim LM, Petrlova J, Schmidtchen A. Cell-free DNA promotes thrombin autolysis and generation of thrombin-derived C-terminal fragments. Front Immunol 2021; 12: 593020
  • 84 Greinacher A, Thiele T, Warkentin TE, Weisser K, Kyrle PA, Eichinger S. Thrombotic thrombocytopenia after ChAdOx1 nCoV-19 vaccination. N Engl J Med 2021; 384 (22) 2092-2101
  • 85 Greinacher A, Selleng K, Palankar R. et al. Insights in ChAdOx1 nCoV-19 vaccine-induced immune thrombotic thrombocytopenia. Blood 2021; 138 (22) 2256-2268
  • 86 Gajsiewicz JM, Smith SA, Morrissey JH. Polyphosphate and RNA differentially modulate the contact pathway of blood clotting. J Biol Chem 2017; 292 (05) 1808-1814
  • 87 Blüm P, Pircher J, Merkle M. et al. Arterial thrombosis in the context of HCV-associated vascular disease can be prevented by protein C. Cell Mol Immunol 2017; 14 (12) 986-996
  • 88 Vogel B, Shinagawa H, Hofmann U, Ertl G, Frantz S. Acute DNase1 treatment improves left ventricular remodeling after myocardial infarction by disruption of free chromatin. Basic Res Cardiol 2015; 110 (02) 15
  • 89 Weber C, Jenke A, Chobanova V. et al. Targeting of cell-free DNA by DNase I diminishes endothelial dysfunction and inflammation in a rat model of cardiopulmonary bypass. Sci Rep 2019; 9 (01) 19249
  • 90 von Brühl ML, Stark K, Steinhart A. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209 (04) 819-835
  • 91 Mai SH, Khan M, Dwivedi DJ. et al; Canadian Critical Care Translational Biology Group. Delayed but not early treatment with DNase reduces organ damage and improves outcome in a murine model of sepsis. Shock 2015; 44 (02) 166-172
  • 92 Vu TT, Zhou J, Leslie BA. et al. Arterial thrombosis is accelerated in mice deficient in histidine-rich glycoprotein. Blood 2015; 125 (17) 2712-2719
  • 93 Kleinert E, Langenmayer MC, Reichart B. et al. Ribonuclease (RNase) prolongs survival of grafts in experimental heart transplantation. J Am Heart Assoc 2016; 5 (05) 5
  • 94 Ohashi A, Murata A, Cho Y. et al. The expression and localization of RNase and RNase inhibitor in blood cells and vascular endothelial cells in homeostasis of the vascular system. PLoS One 2017; 12 (03) e0174237
  • 95 Kluever AK, Deindl E. Extracellular RNA, a potential drug target for alleviating atherosclerosis, ischemia/reperfusion injury and organ transplantation. Curr Pharm Biotechnol 2018; 19 (15) 1189-1195
  • 96 Liang H, Peng B, Dong C. et al. Cationic nanoparticle as an inhibitor of cell-free DNA-induced inflammation. Nat Commun 2018; 9 (01) 4291
  • 97 Fant K, Esbjörner EK, Lincoln P, Nordén B. DNA condensation by PAMAM dendrimers: self-assembly characteristics and effect on transcription. Biochemistry 2008; 47 (06) 1732-1740
  • 98 Utsuno K, Uludağ H. Thermodynamics of polyethylenimine-DNA binding and DNA condensation. Biophys J 2010; 99 (01) 201-207
  • 99 Jain S, Pitoc GA, Holl EK. et al. Nucleic acid scavengers inhibit thrombosis without increasing bleeding. Proc Natl Acad Sci U S A 2012; 109 (32) 12938-12943
  • 100 Lee J, Jackman JG, Kwun J. et al. Nucleic acid scavenging microfiber mesh inhibits trauma-induced inflammation and thrombosis. Biomaterials 2017; 120: 94-102
  • 101 Naqvi I, Giroux N, Olson L. et al. DAMPs/PAMPs induce monocytic TLR activation and tolerance in COVID-19 patients; nucleic acid binding scavengers can counteract such TLR agonists. Biomaterials 2022; 283: 121393
  • 102 Brinkmann V, Reichard U, Goosmann C. et al. Neutrophil extracellular traps kill bacteria. Science 2004; 303 (5663): 1532-1535
  • 103 Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med 2010; 207 (09) 1853-1862
  • 104 Wang Y, Li M, Stadler S. et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 2009; 184 (02) 205-213
  • 105 Neeli I, Khan SN, Radic M. Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol 2008; 180 (03) 1895-1902
  • 106 Papayannopoulos V, Zychlinsky A. NETs: a new strategy for using old weapons. Trends Immunol 2009; 30 (11) 513-521
  • 107 Ermert D, Zychlinsky A, Urban C. Fungal and bacterial killing by neutrophils. In: Rupp S, Sohn K. eds. Host-Pathogen Interactions: Methods and Protocols. Totowa, NJ: Humana Press; 2009: 293-312
  • 108 Hisada Y, Grover SP, Maqsood A. et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020; 105 (01) 218-225
  • 109 Fuchs TA, Kremer Hovinga JA, Schatzberg D, Wagner DD, Lämmle B. Circulating DNA and myeloperoxidase indicate disease activity in patients with thrombotic microangiopathies. Blood 2012; 120 (06) 1157-1164
  • 110 Nakazawa D, Tomaru U, Yamamoto C, Jodo S, Ishizu A. Abundant neutrophil extracellular traps in thrombus of patient with microscopic polyangiitis. Front Immunol 2012; 3: 333
  • 111 de Boer OJ, Li X, Teeling P. et al. Neutrophils, neutrophil extracellular traps and interleukin-17 associate with the organisation of thrombi in acute myocardial infarction. Thromb Haemost 2013; 109 (02) 290-297
  • 112 Riegger J, Byrne RA, Joner M. et al; Prevention of Late Stent Thrombosis by an Interdisciplinary Global European Effort (PRESTIGE) Investigators. Histopathological evaluation of thrombus in patients presenting with stent thrombosis. A multicenter European study: a report of the prevention of late stent thrombosis by an interdisciplinary global European effort consortium. Eur Heart J 2016; 37 (19) 1538-1549
  • 113 Savchenko AS, Martinod K, Seidman MA. et al. Neutrophil extracellular traps form predominantly during the organizing stage of human venous thromboembolism development. J Thromb Haemost 2014; 12 (06) 860-870
  • 114 McDonald B, Davis R, Jenne CN. Neutrophil extracellular traps (NETs) promote disseminated intravascular coagulation in sepsis. J Immunol 2016; 196 (S1): 60-68
  • 115 McDonald B, Davis RP, Kim SJ. et al. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood 2017; 129 (10) 1357-1367
  • 116 Jiménez-Alcázar M, Rangaswamy C, Panda R. et al. Host DNases prevent vascular occlusion by neutrophil extracellular traps. Science 2017; 358 (6367): 1202-1206
  • 117 Noubouossie DF, Whelihan MF, Yu YB. et al. In vitro activation of coagulation by human neutrophil DNA and histone proteins but not neutrophil extracellular traps. Blood 2017; 129 (08) 1021-1029
  • 118 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 (07) 1952-1961
  • 119 Matafonov A, Ivanov IS, Sun M, Gailani D. Coagulation factor XI and factor XII in DNA-induced thrombin generation. Blood 2014; 124: 581
  • 120 Ducroux C, Di Meglio L, Loyau S. et al. Thrombus neutrophil extracellular traps content impair tPA-induced thrombolysis in acute ischemic stroke. Stroke 2018; 49 (03) 754-757
  • 121 Esmon CT. Extracellular histones zap platelets. Blood 2011; 118 (13) 3456-3457
  • 122 Fuchs TA, Bhandari AA, Wagner DD. Histones induce rapid and profound thrombocytopenia in mice. Blood 2011; 118 (13) 3708-3714
  • 123 Gollomp K, Sarkar A, Harikumar S. et al. Fc-modified HIT-like monoclonal antibody as a novel treatment for sepsis. Blood 2020; 135 (10) 743-754
  • 124 Martinod K, Fuchs TA, Zitomersky NL. et al. PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock. Blood 2015; 125 (12) 1948-1956