Understanding Infection-Induced Thrombosis: Lessons Learned From Animal Models

Nonantzin Beristain-Covarrubias, Marisol Perez-Toledo, Mark R Thomas, Ian R Henderson, Steve P Watson, Adam F Cunningham, Nonantzin Beristain-Covarrubias, Marisol Perez-Toledo, Mark R Thomas, Ian R Henderson, Steve P Watson, Adam F Cunningham

Abstract

Thrombosis is a common consequence of infection that is associated with poor patient outcome. Nevertheless, the mechanisms by which infection-associated thrombosis is induced, maintained and resolved are poorly understood, as is the contribution thrombosis makes to host control of infection and pathogen spread. The key difference between infection-associated thrombosis and thrombosis in other circumstances is a stronger inflammation-mediated component caused by the presence of the pathogen and its products. This inflammation triggers the activation of platelets, which may accompany damage to the endothelium, resulting in fibrin deposition and thrombus formation. This process is often referred to as thrombo-inflammation. Strikingly, despite its clinical importance and despite thrombi being induced to many different pathogens, it is still unclear whether the mechanisms underlying this process are conserved and how we can best understand this process. This review summarizes thrombosis in a variety of models, including single antigen models such as LPS, and infection models using viruses and bacteria. We provide a specific focus on Salmonella Typhimurium infection as a useful model to address all stages of thrombosis during infection. We highlight how this model has helped us identify how thrombosis can appear in different organs at different times and thrombi be detected for weeks after infection in one site, yet largely be resolved within 24 h in another. Furthermore, we discuss the observation that thrombi induced to Salmonella Typhimurium are largely devoid of bacteria. Finally, we discuss the value of different therapeutic approaches to target thrombosis, the potential importance of timing in their administration and the necessity to maintain normal hemostasis after treatment. Improvements in our understanding of these processes can be used to better target infection-mediated mechanisms of thrombosis.

Keywords: Salmonella; bacteria; pathogens; platelets; thrombo-inflammation; thrombosis; virus.

Copyright © 2019 Beristain-Covarrubias, Perez-Toledo, Thomas, Henderson, Watson and Cunningham.

Figures

Figure 1
Figure 1
Examples of animal models available to study thrombosis during infection. A range of approaches has been employed to evaluate infection-induced thrombosis. Single microbial component-induced sepsis, or CLP models, mimic severe sepsis in humans, but most are usually only useable for a few days either because the infection is cleared or it is lethal. Some viral and bacterial infection models can induce longer-lasting infections and thrombotic complications may also be useful to study the resolution of thrombosis.
Figure 2
Figure 2
Thrombosis develops asynchronously in the spleen and liver after STm infection. Resident immune cells (monocytes/macrophages/neutrophils) are more abundant in the spleen compared to the liver. Although the mechanism is still under study, splenic phagocytic cells and neutrophils participate in the induction of white, platelet-rich, thrombi in the spleen 24 h after infection. Thrombi in the spleen resolve rapidly thereafter so that from day 7 onwards post-infection only “ghosts” of fibrin are observed. In the liver, thrombi are not detected until 7 days after infection, despite the liver and spleen having similarly high bacteria burdens in the first week post-infection. Thrombosis in the liver requires inflammatory cell recruitment and activation through TLR4, which promotes IFNγ production, the accumulation of monocytic cells and their upregulation of podoplanin. Inflammation is associated with perturbed endothelial integrity/endothelial damage making it possible for the interaction of podoplanin expressed on monocytic cells and CLEC-2 on platelets. Thrombosis in the liver persists for weeks and the resolution of thrombosis is observed from day 21 after infection. The mechanism(s) that underlie the resolution of thrombosis in these two different organs is still unknown. A strength of the STm infection model is that it offers an alternative tool to understand this process.
Figure 3
Figure 3
Current and potential molecules or pathways to target infection-associated thrombosis. Anticoagulants interfere with the activity of different clotting factors in the coagulation cascade that aims to rebalance hemostasis. Platelet GPVI and CLEC-2 are activated via SFK kinases and play a role in infection-mediated platelet activation. Neutrophil activation and the release of NETs together with associated factors (DNA/Histones) are also under study for developing strategies to limit their pro-thrombotic effects.

References

    1. 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) 367:1075–9. 10.1016/S0140-6736(06)68474-2
    1. Dalager-Pedersen M, Sogaard M, Schonheyder HC, Nielsen H, Thomsen RW. Risk for myocardial infarction and stroke after community-acquired bacteremia: a 20-year population-based cohort study. Circulation. (2014) 129:1387–96. 10.1161/CIRCULATIONAHA.113.006699
    1. Cohoon KP, Ashrani AA, Crusan DJ, Petterson TM, Bailey KR, Heit JA. Is infection an independent risk factor for venous thromboembolism? A population-based, case-control study. Am J Med. (2018) 131:307–16.e2. 10.1016/j.amjmed.2017.09.015
    1. Liu X, Wang L, Wang S, Zhang W, Yu Y, Chen S, Ao H. Association between infection and thrombosis after coronary artery bypass grafting: a cohort study. J Cardiothorac Vasc Anesth. (2019) 33:1610–6. 10.1053/j.jvca.2018.09.008
    1. Fugate JE, Lyons JL, Thakur KT, Smith BR, Hedley-Whyte ET, Mateen FJ. Infectious causes of stroke. Lancet Infect Dis. (2014) 14:869–80. 10.1016/S1473-3099(14)70755-8
    1. Huang J. Infection and thrombosis in cardiac surgery: is there a common ground? J Cardiothorac Vasc Anesth. (2019) 33: 1617–9. 10.1053/j.jvca.2018.10.003
    1. Dolapsakis C, Kranidioti E, Katsila S, Samarkos M. Cavernous sinus thrombosis due to ipsilateral sphenoid sinusitis. BMJ Case Rep. (2019) 12:e227302. 10.1136/bcr-2018-227302
    1. Oliveira GN, Basso S, Sevivas T, Neves N. Varicella complicated by cellulitis and deep vein thrombosis. BMJ Case Rep. (2017) 2017:bcr-2017-221499. 10.1136/bcr-2017-221499
    1. Kohler JA, Munoz FM, Goss JA, Miloh TA. Viral upper respiratory infection at pediatric liver transplantation is associated with hepatic artery thrombosis. Liver Transpl. (2017) 23:1477–81. 10.1002/lt.24866
    1. Ceccarelli M, Venanzi Rullo E, Nunnari G. Risk factors of venous thrombo-embolism during cytomegalovirus infection in immunocompetent individuals. A systematic review. Eur J Clin Microbiol Infect Dis. (2018) 37:381–90. 10.1007/s10096-018-3185-y
    1. Edula RG, Qureshi K, Khallafi H. Acute cytomegalovirus infection in liver transplant recipients: An independent risk for venous thromboembolism. World J Hepatol. (2013) 5:692–5. 10.4254/wjh.v5.i12.692
    1. Samarasekara K, Munasinghe J. Dengue shock syndrome complicated with acute liver failure and kidney injury, infective endocarditis, and deep vein thrombosis: a case report. J Med Case Rep. (2018) 12:321. 10.1186/s13256-018-1862-1
    1. Roquer J, Cuadrado-Godia E, Giralt-Steinthauer E, Jimena S, Jimenez-Conde J, Martinez-Rodriguez JE, et al. . Previous infection and stroke: a prospective study. Cerebrovasc Dis. (2012) 33:310–5. 10.1159/000335306
    1. Corrales-Medina VF, Madjid M, Musher DM. Role of acute infection in triggering acute coronary syndromes. Lancet Infect Dis. (2010) 10:83–92. 10.1016/S1473-3099(09)70331-7
    1. Leinonen M, Saikku P. Evidence for infectious agents in cardiovascular disease and atherosclerosis. Lancet Infect Dis. (2002) 2:11–7. 10.1016/S1473-3099(01)00168-2
    1. Musher DM, Abers MS, Corrales-Medina VF. Acute infection and myocardial infarction. N Engl J Med. (2019) 380:171–6. 10.1056/NEJMra1808137
    1. Franchi T, Eaton S, De Coppi P, Giuliani S. The emerging role of immunothrombosis in paediatric conditions. Pediatr Res. (2019) 86:19–27. 10.1038/s41390-019-0343-6
    1. Ferguson JH, Chapman OD. Fulminating meningococcic infections and the so-called Waterhouse-Friderichsen syndrome. Am J Pathol. (1948) 24:763–95.
    1. Margaretten W, Mc AA. An appraisal of fulminant meningococcemia with reference to the Shartzman phenomenon. Am J Med. (1958) 25:868–76. 10.1016/0002-9343(58)90059-7
    1. Heyderman RS. Sepsis and intravascular thrombosis. Arch Dis Child. (1993) 68:621–3. 10.1136/adc.68.5.621
    1. Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov. (2005) 4:854–65. 10.1038/nrd1854
    1. Ho LWW, Kam P, Thong CL. Disseminated intravascular coagulation. Curr Anesth Critic Care. (2005) 16:151–61. 10.1016/j.cacc.2005.03.011
    1. Tani VM, Assis-Mendonca GR, da Silva TB, Rogerio F, De Paula EV. Microvascular thrombosis in sepsis: an autopsy study. Thromb Res. (2017) 156:23–5. 10.1016/j.thromres.2017.05.031
    1. Mauriello A, Sangiorgi G, Fratoni S, Palmieri G, Bonanno E, Anemona L, et al. . Diffuse and active inflammation occurs in both vulnerable and stable plaques of the entire coronary tree: a histopathologic study of patients dying of acute myocardial infarction. J Am Coll Cardiol. (2005) 45:1585–93. 10.1016/j.jacc.2005.01.054
    1. Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, et al. . Extracellular histones are major mediators of death in sepsis. Nat Med. (2009) 15:1318–21. 10.1038/nm.2053
    1. Lahteenmaki K, Kukkonen M, Korhonen TK. The Pla surface protease/adhesin of Yersinia pestis mediates bacterial invasion into human endothelial cells. FEBS Lett. (2001) 504:69–72. 10.1016/S0014-5793(01)02775-2
    1. Coleman JL, Gebbia JA, Piesman J, Degen JL, Bugge TH, Benach JL. Plasminogen is required for efficient dissemination of B. burgdorferi in ticks and for enhancement of spirochetemia in mice. Cell. (1997) 89:1111–9. 10.1016/S0092-8674(00)80298-6
    1. Lahteenmaki K, Kuusela P, Korhonen TK. Bacterial plasminogen activators and receptors. FEMS Microbiol Rev. (2001) 25:531–52. 10.1111/j.1574-6976.2001.tb00590.x
    1. Lee WY, Moriarty TJ, Wong CH, Zhou H, Strieter RM, van Rooijen N, et al. . An intravascular immune response to Borrelia burgdorferi involves Kupffer cells and iNKT cells. Nat Immunol. (2010) 11:295–302. 10.1038/ni.1855
    1. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, Jr, et al. . Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. (2010) 107:15880–5. 10.1073/pnas.1005743107
    1. Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S, Goosmann C, et al. . Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. (2010) 16:887–96. 10.1038/nm.2184
    1. Berthelsen LO, Kristensen AT, Tranholm M. Animal models of DIC and their relevance to human DIC: a systematic review. Thromb Res. (2011) 128:103–16. 10.1016/j.thromres.2010.12.002
    1. Cohen J. The immunopathogenesis of sepsis. Nature. (2002) 420:885–91. 10.1038/nature01326
    1. Pawlinski R, Pedersen B, Schabbauer G, Tencati M, Holscher T, Boisvert W, et al. . Role of tissue factor and protease-activated receptors in a mouse model of endotoxemia. Blood. (2004) 103:1342–7. 10.1182/blood-2003-09-3051
    1. Yanada M, Kojima T, Ishiguro K, Nakayama Y, Yamamoto K, Matsushita T, et al. . Impact of antithrombin deficiency in thrombogenesis: lipopolysaccharide and stress-induced thrombus formation in heterozygous antithrombin-deficient mice. Blood. (2002) 99:2455–8. 10.1182/blood.V99.7.2455
    1. Hermida J, Montes R, Paramo JA, Rocha E. Endotoxin-induced disseminated intravascular coagulation in rabbits: effect of recombinant hirudin on hemostatic parameters, fibrin deposits, and mortality. J Lab Clin Med. (1998) 131:77–83. 10.1016/S0022-2143(98)90080-4
    1. Levi M, Dorffler-Melly J, Reitsma P, Buller H, Florquin S, van der Poll T, Carmeliet P. Aggravation of endotoxin-induced disseminated intravascular coagulation and cytokine activation in heterozygous protein-C-deficient mice. Blood. (2003) 101:4823–7. 10.1182/blood-2002-10-3254
    1. Xiang B, Zhang G, Guo L, Li XA, Morris AJ, Daugherty A, et al. . Platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the cyclooxygenase 1 signalling pathway. Nat Commun. (2013) 4:2657. 10.1038/ncomms3657
    1. Armstrong MT, Rickles FR, Armstrong PB. Capture of lipopolysaccharide (endotoxin) by the blood clot: a comparative study. PLoS ONE. (2013) 8:e80192. 10.1371/journal.pone.0080192
    1. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol. (2013) 13:34–45. 10.1038/nri3345
    1. Berube BJ, Bubeck Wardenburg J. Staphylococcus aureus α-toxin: nearly a century of intrigue. Toxins (Basel). (2013) 5:1140–66. 10.3390/toxins5061140
    1. Parimon T, Li Z, Bolz DD, McIndoo ER, Bayer CR, Stevens DL, et al. . Staphylococcus aureus α-hemolysin promotes platelet-neutrophil aggregate formation. J Infect Dis. (2013) 208:761–70. 10.1093/infdis/jit235
    1. Surewaard BGJ, Thanabalasuriar A, Zeng Z, Tkaczyk C, Cohen TS, Bardoel BW, et al. . α-Toxin induces platelet aggregation and liver injury during staphylococcus aureus sepsis. Cell Host Microbe. (2018) 24:271–84.e3. 10.1016/j.chom.2018.06.017
    1. Fortier ME, Kent S, Ashdown H, Poole S, Boksa P, Luheshi GN. The viral mimic, polyinosinic:polycytidylic acid, induces fever in rats via an interleukin-1-dependent mechanism. Am J Physiol Regul Integr Comp Physiol. (2004) 287:R759–66. 10.1152/ajpregu.00293.2004
    1. Antoniak S, Tatsumi K, Bode M, Vanja S, Williams JC, Mackman N. Protease-activated receptor 1 enhances poly i:c induction of the antiviral response in macrophages and mice. J Innate Immun. (2017) 9:181–92. 10.1159/000450853
    1. D'Atri LP, Etulain J, Rivadeneyra L, Lapponi MJ, Centurion M, Cheng K, et al. . Expression and functionality of Toll-like receptor 3 in the megakaryocytic lineage. J Thromb Haemost. (2015) 13:839–50. 10.1111/jth.12842
    1. Blum P, Pircher J, Merkle M, Czermak T, Ribeiro A, Mannell H, et al. . Arterial thrombosis in the context of HCV-associated vascular disease can be prevented by protein C. Cell Mol Immunol. (2017) 14:986–96. 10.1038/cmi.2016.10
    1. Levi M, Ten Cate H. Disseminated intravascular coagulation. N Engl J Med. (1999) 341:586–92. 10.1056/NEJM199908193410807
    1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. (2013) 369:840–51. 10.1056/NEJMra1208623
    1. Fay KT, Klingensmith NJ, Chen CW, Zhang W, Sun Y, Morrow KN, et al. . The gut microbiome alters immunophenotype and survival from sepsis. FASEB J. (2019) 33:11258–69. 10.1096/fj.201802188R
    1. Inoue Y, Kohno S, Miyazaki T, Yamaguchi K. Effect of a platelet activating factor antagonist and antithrombin III on septicemia and endotoxemia in rats. Tohoku J Exp Med. (1991) 163:175–85. 10.1620/tjem.163.175
    1. Rayes J, Lax S, Wichaiyo S, Watson SK, Di Y, Lombard S, et al. . The podoplanin-CLEC-2 axis inhibits inflammation in sepsis. Nat Commun. (2017) 8:2239. 10.1038/s41467-017-02402-6
    1. Vardon Bounes F, Memier V, Marcaud M, Jacquemin A, Hamzeh-Cognasse H, Garcia C, et al. . Platelet activation and prothrombotic properties in a mouse model of peritoneal sepsis. Sci Rep. (2018) 8:13536. 10.1038/s41598-018-31910-8
    1. Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-interactions: linking hemostasis and inflammation. Blood Rev. (2007) 21:99–111. 10.1016/j.blre.2006.06.001
    1. Wang Z, Su F, Rogiers P, Vincent JL. Beneficial effects of recombinant human activated protein C in a ewe model of septic shock. Crit Care Med. (2007) 35:2594–600. 10.1097/01.CCM.0000287590.55294.40
    1. Wigton DH, Kociba GJ, Hoover EA. Infectious canine hepatitis: animal model for viral-induced disseminated intravascular coagulation. Blood. (1976) 47:287–96. 10.1182/blood.V47.2.287.bloodjournal472287
    1. Clar C, Oseni Z, Flowers N, Keshtkar-Jahromi M, Rees K. Influenza vaccines for preventing cardiovascular disease. Cochrane Database Syst Rev. (2015) CD005050. 10.1002/14651858.CD005050.pub3 [Epub ahead of print].
    1. Linder M, Muller-Berghaus G, Lasch HG, Gagel C. Virus infection and blood coagulation. Thromb Diath Haemorrh. (1970) 23:1–11.
    1. Yang Y, Tang H. Aberrant coagulation causes a hyper-inflammatory response in severe influenza pneumonia. Cell Mol Immunol. (2016) 13:432–42. 10.1038/cmi.2016.1
    1. Cui S, Fu Z, Feng Y, Xie X, Ma X, Liu T, et al. . The disseminated intravascular coagulation score is a novel predictor for portal vein thrombosis in cirrhotic patients with hepatitis B. Thromb Res. (2018) 161:7–11. 10.1016/j.thromres.2017.11.010
    1. Geisbert TW, Young HA, Jahrling PB, Davis KJ, Kagan E, Hensley LE. Mechanisms underlying coagulation abnormalities in ebola hemorrhagic fever: overexpression of tissue factor in primate monocytes/macrophages is a key event. J Infect Dis. (2003) 188:1618–29. 10.1086/379724
    1. Schechter ME, Andrade BB, He T, Richter GH, Tosh KW, Policicchio BB, et al. . Inflammatory monocytes expressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med. (2017) 9:eaam5441. 10.1126/scitranslmed.aam5441
    1. Short KR, Kroeze E, Fouchier RAM, Kuiken T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect Dis. (2014) 14:57–69. 10.1016/S1473-3099(13)70286-X
    1. Le VB, Schneider JG, Boergeling Y, Berri F, Ducatez M, Guerin JL, et al. . Platelet activation and aggregation promote lung inflammation and influenza virus pathogenesis. Am J Respir Crit Care Med. (2015) 191:804–19. 10.1164/rccm.201406-1031OC
    1. Ashar HK, Mueller NC, Rudd JM, Snider TA, Achanta M, Prasanthi M, et al. . The role of extracellular histones in influenza virus pathogenesis. Am J Pathol. (2018) 188:135–48. 10.1016/j.ajpath.2017.09.014
    1. Yacoub D, Hachem A, Theoret JF, Gillis MA, Mourad W, Merhi Y. Enhanced levels of soluble CD40 ligand exacerbate platelet aggregation and thrombus formation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway. Arterioscler Thromb Vasc Biol. (2010) 30:2424–33. 10.1161/ATVBAHA.110.216143
    1. Kipnis E, Guery BP, Tournoys A, Leroy X, Robriquet L, Fialdes P, et al. . Massive alveolar thrombin activation in Pseudomonas aeruginosa-induced acute lung injury. Shock. (2004) 21:444–51. 10.1097/00024382-200405000-00008
    1. Renckens R, Roelofs JJ, Stegenga ME, Florquin S, Levi M, Carmeliet P, et al. . Transgenic tissue-type plasminogen activator expression improves host defense during Klebsiella pneumonia. J Thromb Haemost. (2008) 6:660–8. 10.1111/j.1538-7836.2008.02892.x
    1. Skjeflo EW, Christiansen D, Fure H, Ludviksen JK, Woodruff TM, Espevik T, et al. . Staphylococcus aureus-induced complement activation promotes tissue factor-mediated coagulation. J Thromb Haemost. (2018) 16:905–18. 10.1111/jth.13979
    1. Korhonen TK, Haiko J, Laakkonen L, Jarvinen HM, Westerlund-Wikstrom B. Fibrinolytic and coagulative activities of Yersinia pestis. Front Cell Infect Microbiol. (2013) 3:35. 10.3389/fcimb.2013.00035
    1. Sun H, Ringdahl U, Homeister JW, Fay WP, Engleberg NC, Yang AY, et al. . Plasminogen is a critical host pathogenicity factor for group A streptococcal infection. Science. (2004) 305:1283–6. 10.1126/science.1101245
    1. Luo D, Szaba FM, Kummer LW, Plow EF, Mackman N, Gailani D, et al. Protective roles for fibrin, tissue factor, plasminogen activator inhibitor-1, and thrombin activatable fibrinolysis inhibitor, but not factor XI, during defense against the gram-negative bacterium Yersinia enterocolitica. J Immunol. (2011) 187:1866–76. 10.4049/jimmunol.1101094
    1. Popova TG, Millis B, Bailey C, Popov SG. Platelets, inflammatory cells, von Willebrand factor, syndecan-1, fibrin, fibronectin, and bacteria co-localize in the liver thrombi of Bacillus anthracis-infected mice. Microb Pathog. (2012) 52:1–9. 10.1016/j.micpath.2011.08.004
    1. Hitchcock JR, Cook CN, Bobat S, Ross EA, Flores-Langarica A, Lowe KL, et al. . Inflammation drives thrombosis after Salmonella infection via CLEC-2 on platelets. J Clin Invest. (2015) 125:4429–46. 10.1172/JCI79070
    1. Wong CH, Jenne CN, Petri B, Chrobok NL, Kubes P. Nucleation of platelets with blood-borne pathogens on Kupffer cells precedes other innate immunity and contributes to bacterial clearance. Nat Immunol. (2013) 14:785–92. 10.1038/ni.2631
    1. Robriquet L, Collet F, Tournoys A, Prangere T, Neviere R, Fourrier F, et al. . Intravenous administration of activated protein C in Pseudomonas-induced lung injury: impact on lung fluid balance and the inflammatory response. Respir Res. (2006) 7:41. 10.1186/1465-9921-7-41
    1. Choi G, Hofstra JJ, Roelofs JJ, Florquin S, Bresser P, Levi M, et al. . Recombinant human activated protein C inhibits local and systemic activation of coagulation without influencing inflammation during Pseudomonas aeruginosa pneumonia in rats. Crit Care Med. (2007) 35:1362–8. 10.1097/01.CCM.0000261888.32654.6D
    1. Matt U, Warszawska JM, Bauer M, Dietl W, Mesteri I, Doninger B, et al. . Bβ(15-42) protects against acid-induced acute lung injury and secondary pseudomonas pneumonia in vivo. Am J Respir Crit Care Med. (2009) 180:1208–17. 10.1164/rccm.200904-0626OC
    1. Wollein Waldetoft K, Mohanty T, Karlsson C, Morgelin M, Frick IM, Malmstrom J, et al. . Saliva-induced clotting captures streptococci: novel roles for coagulation and fibrinolysis in host defense and immune evasion. Infect Immun. (2016) 84:2813–23. 10.1128/IAI.00307-16
    1. Sun H, Wang X, Degen JL, Ginsburg D. Reduced thrombin generation increases host susceptibility to group A streptococcal infection. Blood. (2009) 113:1358–64. 10.1182/blood-2008-07-170506
    1. Kahn F, Hurley S, Shannon O. Platelets promote bacterial dissemination in a mouse model of streptococcal sepsis. Microbes Infect. (2013) 15:669–76. 10.1016/j.micinf.2013.05.003
    1. Pahlman LI, Morgelin M, Kasetty G, Olin AI, Schmidtchen A, Herwald H. Antimicrobial activity of fibrinogen and fibrinogen-derived peptides–a novel link between coagulation and innate immunity. Thromb Haemost. (2013) 109:930–9. 10.1160/TH12-10-0739
    1. Macrae FL, Duval C, Papareddy P, Baker SR, Yuldasheva N, Kearney KJ, et al. . A fibrin biofilm covers blood clots and protects from microbial invasion. J Clin Invest. (2018) 128:3356–68. 10.1172/JCI98734
    1. Brown DE, McCoy MW, Pilonieta MC, Nix RN, Detweiler CS. Chronic murine typhoid fever is a natural model of secondary hemophagocytic lymphohistiocytosis. PLoS ONE. (2010) 5:e9441. 10.1371/journal.pone.0009441
    1. Hickey MJ, Kubes P. Intravascular immunity: the host-pathogen encounter in blood vessels. Nat Rev Immunol. (2009) 9:364–75. 10.1038/nri2532
    1. Feasey NA, Dougan G, Kingsley RA, Heyderman RS, Gordon MA. Invasive non-typhoidal salmonella disease: an emerging and neglected tropical disease in Africa. Lancet. (2012) 379:2489–99. 10.1016/S0140-6736(11)61752-2
    1. Wright AE, Knapp HH. A note on the causation and treatment of thrombosis occurring in connection with typhoid fever. Med Chir Trans. (1903) 86:1–19.
    1. Huckstep RL. Typhoid Fever and other Salmonella Infections, E. & Livingstone S. BJS Soc. (1962) 51:238 10.1002/bjs.1800510322
    1. Mohanty S, Bakshi S, Gupta AK, Kapil A, Arya LS, Das BK. Venous thrombosis associated with Salmonella: report of a case and review of literature. Indian J Med Sci. (2003) 57:199–203.
    1. Schifferdecker B, Merchan JA, Ahmar C, Worthington M, Griben A, Schainfeld RM, et al. . Endovascular treatment of septic thrombophlebitis: a case report of a rare complication and review of the literature. Vasc Med. (2009) 14:47–50. 10.1177/1358863X08096517
    1. Ceyhan M, Kanra G, Benderlioglu B, Secmeer G, Hicsonmez G, Kirazli S. Transient protein S deficiency with deep venous thrombosis during Salmonella Typhimurium infection. Arch Dis Child. (1993) 68:138–9. 10.1136/adc.68.1.138
    1. Salamon SA, Prag J. A case of superficial septic thrombophlebitis in a varicose vein caused by Salmonella panama. Clin Microbiol Infect. (2001) 7:34–6. 10.1046/j.1469-0691.2001.00182.x
    1. Carey J, Buchstein S, Shah S. Septic deep vein thrombosis due to Salmonella johannesburg. J Infect. (2001) 42:79–80. 10.1053/jinf.2000.0762
    1. Uribe JAZ, Coura FM, Nunes P, Silva MVP, de Carvalho AU, Moreira MVL, et al. Septicemic salmonellosis in pre weaned calves caused by Salmonella dublin. Res J Vet Pract. (2015) 3:69–75. 10.14737/journal.rjvp/2015/3.3.69.75
    1. Latour JG, Leger C, Renaud S, Simard P. On the mechanisms responsible for selection of hepatic veins as target for thrombosis following injection of endotoxin in hyperlipemic rats. Am J Pathol. (1974) 76:195–212.
    1. Innes JR, Wilson C, Ross MA. Epizootic Salmonella enteritidis infection causing septic pulmonary phlebothrombosis in hamsters. J Infect Dis. (1956) 98:133–41. 10.1093/infdis/98.2.133
    1. Loomis WP, Johnson ML, Brasfield A, Blanc MP, Yi J, Miller SI, et al. . Temporal and anatomical host resistance to chronic Salmonella infection is quantitatively dictated by Nramp1 and influenced by host genetic background. PLoS ONE. (2014) 9:e111763. 10.1371/journal.pone.0111763
    1. Vidal SM, Pinner E, Lepage P, Gauthier S, Gros P. Natural resistance to intracellular infections: Nramp1 encodes a membrane phosphoglycoprotein absent in macrophages from susceptible (Nramp1 D169) mouse strains. J Immunol. (1996) 157:3559–68.
    1. Monack DM, Bouley DM, Falkow S. Salmonella Typhimurium persists within macrophages in the mesenteric lymph nodes of chronically infected Nramp1+/+ mice and can be reactivated by IFNgamma neutralization. J Exp Med. (2004) 199:231–41. 10.1084/jem.20031319
    1. Hoiseth SK, Stocker BA. Aromatic-dependent Salmonella Typhimurium are non-virulent and effective as live vaccines. Nature. (1981) 291:238–9. 10.1038/291238a0
    1. Cunningham AF, Gaspal F, Serre K, Mohr E, Henderson IR, Scott-Tucker A, et al. . Salmonella induces a switched antibody response without germinal centers that impedes the extracellular spread of infection. J Immunol. (2007) 178:6200–7. 10.4049/jimmunol.178.10.6200
    1. Ross EA, Coughlan RE, Flores-Langarica A, Bobat S, Marshall JL, Hussain K, et al. . CD31 is required on CD4+ T cells to promote T cell survival during Salmonella infection. J Immunol. (2011) 187:1553–65. 10.4049/jimmunol.1000502
    1. Bobat S, Darby M, Mrdjen D, Cook C, Logan E, Auret J, et al. . Natural and vaccine-mediated immunity to Salmonella Typhimurium is impaired by the helminth Nippostrongylus brasiliensis. PLoS Negl Trop Dis. (2014) 8:e3341. 10.1371/journal.pntd.0003341
    1. Beristain-Covarrubias N, Perez-Toledo M, Flores-Langarica A, Zuidscherwoude M, Hitchcock JR, Channell WM, et al. Salmonella-induced thrombi in mice develop asynchronously in the spleen and liver and are not effective bacterial traps. Blood. (2019) 133:600–4. 10.1182/blood-2018-08-867267
    1. Tam JW, Kullas AL, Mena P, Bliska JB, van der Velden AW. CD11b+ Ly6Chi Ly6G- immature myeloid cells recruited in response to Salmonella enterica serovar Typhimurium infection exhibit protective and immunosuppressive properties. Infect Immun. (2014) 82:2606–14. 10.1128/IAI.01590-13
    1. Sheppard M, Webb C, Heath F, Mallows V, Emilianus R, Maskell D, et al. . Dynamics of bacterial growth and distribution within the liver during Salmonella infection. Cell Microbiol. (2003) 5:593–600. 10.1046/j.1462-5822.2003.00296.x
    1. Conlan JW, North RJ. Listeria monocytogenes, but not Salmonella Typhimurium, elicits a CD18-independent mechanism of neutrophil extravasation into the murine peritoneal cavity. Infect Immun. (1994) 62:2702–6.
    1. Gonzalez-Escobedo G, La Perle KM, Gunn JS. Histopathological analysis of Salmonella chronic carriage in the mouse hepatopancreatobiliary system. PLoS ONE. (2013) 8:e84058. 10.1371/journal.pone.0084058
    1. Erova TE, Kirtley ML, Fitts EC, Ponnusamy D, Baze WB, Andersson JA, et al. . Protective immunity elicited by oral immunization of mice with Salmonella enterica Serovar Typhimurium Braun Lipoprotein (Lpp) and Acetyltransferase (MsbB) mutants. Front Cell Infect Microbiol. (2016) 6:148. 10.3389/fcimb.2016.00148
    1. Wickham ME, Brown NF, Provias J, Finlay BB, Coombes BK. Oral infection of mice with Salmonella enterica serovar Typhimurium causes meningitis and infection of the brain. BMC Infect Dis. (2007) 7:65. 10.1186/1471-2334-7-65
    1. Ross EA, Coughlan RE, Flores-Langarica A, Lax S, Nicholson J, Desanti GE, et al. . Thymic function is maintained during Salmonella-induced atrophy and recovery. J Immunol. (2012) 189:4266–74. 10.4049/jimmunol.1200070
    1. Ross EA, Flores-Langarica A, Bobat S, Coughlan RE, Marshall JL, Hitchcock JR, et al. . Resolving Salmonella infection reveals dynamic and persisting changes in murine bone marrow progenitor cell phenotype and function. Eur J Immunol. (2014) 44:2318–30. 10.1002/eji.201344350
    1. Suzuki-Inoue K, Fuller GL, Garcia A, Eble JA, Pohlmann S, Inoue O, et al. . A novel Syk-dependent mechanism of platelet activation by the C-type lectin receptor CLEC-2. Blood. (2006) 107:542–9. 10.1182/blood-2005-05-1994
    1. Finegold MJ. Pathogenesis of plague. A review of plague deaths in the United States during the last decade. Am J Med. (1968) 45:549–54. 10.1016/0002-9343(68)90171-X
    1. Lucas S. The autopsy pathology of sepsis-related death. Curr Diagnost Pathol. (2007) 13:375–88. 10.1016/j.cdip.2007.06.001
    1. Tan KT, Lip GY. Red vs white thrombi: treating the right clot is crucial. Arch Intern Med. (2003) 163:2534–5; author reply 2535. 10.1001/archinte.163.20.2534-b
    1. Saracco P, Vitale P, Scolfaro C, Pollio B, Pagliarino M, Timeus F. The coagulopathy in sepsis: significance and implications for treatment. Pediatr Rep. (2011) 3:e30. 10.4081/pr.2011.e30
    1. Allen KS, Sawheny E, Kinasewitz GT. Anticoagulant modulation of inflammation in severe sepsis. World J Crit Care Med. (2015) 4:105–15. 10.5492/wjccm.v4.i2.105
    1. Davis RP, Miller-Dorey S, Jenne CN. Platelets and coagulation in infection. Clin Transl Immunol. (2016) 5:e89. 10.1038/cti.2016.39
    1. Yaghi S, Eisenberger A, Willey JZ. Symptomatic intracerebral hemorrhage in acute ischemic stroke after thrombolysis with intravenous recombinant tissue plasminogen activator: a review of natural history and treatment. JAMA Neurol. (2014) 71:1181–5. 10.1001/jamaneurol.2014.1210
    1. Akol H, Boon E, van Haren F, van der Hoeven J. Successful treatment of fulminant pneumococcal sepsis with recombinant tissue plasminogen activator. Eur J Intern Med. (2002) 13:389. 10.1016/S0953-6205(02)00095-X
    1. Zenz W, Muntean W, Gallistl S, Zobel G, Grubbauer HM. Recombinant tissue plasminogen activator treatment in two infants with fulminant meningococcemia. Pediatrics. (1995) 96:144–8.
    1. Zenz W, Muntean W, Zobel G, Grubbauer HM, Gallistl S. Treatment of fulminant meningococcemia with recombinant tissue plasminogen activator. Thromb Haemost. (1995) 74:802–3. 10.1055/s-0038-1649821
    1. Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, et al. . Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA. (2001) 286:1869–78. 10.1001/jama.286.15.1869
    1. Abraham E, Reinhart K, Opal S, Demeyer I, Doig C, Rodriguez AL, et al. . Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled trial. JAMA. (2003) 290:238–47. 10.1001/jama.290.2.238
    1. Saito H, Maruyama I, Shimazaki S, Yamamoto Y, Aikawa N, Ohno R, et al. . Efficacy and safety of recombinant human soluble thrombomodulin (ART-123) in disseminated intravascular coagulation: results of a phase III, randomized, double-blind clinical trial. J Thromb Haemost. (2007) 5:31–41. 10.1111/j.1538-7836.2006.02267.x
    1. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A, et al. . Recombinant human protein, efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. (2001) 344:699–709. 10.1056/NEJM200103083441001
    1. Zenz W, Zoehrer B, Levin M, Fanconi S, Hatzis TD, Knight G, et al. . Use of recombinant tissue plasminogen activator in children with meningococcal purpura fulminans: a retrospective study. Crit Care Med. (2004) 32:1777–80. 10.1097/01.CCM.0000133667.86429.5D
    1. Jaimes F, De La Rosa G, Morales C, Fortich F, Arango C, Aguirre D, et al. . Unfractioned heparin for treatment of sepsis: a randomized clinical trial (The HETRASE Study). Crit Care Med. (2009) 37:1185–96. 10.1097/CCM.0b013e31819c06bc
    1. Warnock LB, Huang D. Heparin. Treasure Island, FL: StatPearls; (2019).
    1. Fuchs TA, Bhandari AA, Wagner DD. Histones induce rapid and profound thrombocytopenia in mice. Blood. (2011) 118:3708–14. 10.1182/blood-2011-01-332676
    1. Ma AC, Kubes P. Platelets, neutrophils, and neutrophil extracellular traps (NETs) in sepsis. J Thromb Haemost. (2008) 6:415–20. 10.1111/j.1538-7836.2007.02865.x
    1. Jenne CN, Wong CH, Zemp FJ, McDonald B, Rahman MM, Forsyth PA, et al. . Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe. (2013) 13:169–80. 10.1016/j.chom.2013.01.005
    1. Claushuis TAM, de Vos AF, Nieswandt B, Boon L, Roelofs J, de Boer OJ, et al. . Platelet glycoprotein VI aids in local immunity during pneumonia-derived sepsis caused by gram-negative bacteria. Blood. (2018) 131:864–76. 10.1182/blood-2017-06-788067
    1. Lax S, Rayes J, Wichaiyo S, Haining EJ, Lowe K, Grygielska B, et al. . Platelet CLEC-2 protects against lung injury via effects of its ligand podoplanin on inflammatory alveolar macrophages in the mouse. Am J Physiol Lung Cell Mol Physiol. (2017) 313:L1016–29. 10.1152/ajplung.00023.2017
    1. Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest. (2019) 129:12–23. 10.1172/JCI122955
    1. Astarita JL, Acton SE, Turley SJ. Podoplanin: emerging functions in development, the immune system, and cancer. Front Immunol. (2012) 3:283. 10.3389/fimmu.2012.00283
    1. Payne H, Ponomaryov T, Watson SP, Brill A. Mice with a deficiency in CLEC-2 are protected against deep vein thrombosis. Blood. (2017) 129:2013–20. 10.1182/blood-2016-09-742999
    1. Lax S, Rayes J, Thickett DR, Watson SP. Effect of anti-podoplanin antibody administration during lipopolysaccharide-induced lung injury in mice. BMJ Open Respir Res. (2017) 4:e000257. 10.1136/bmjresp-2017-000257
    1. Harrison MJ, Chimen M, Hussain M, Iqbal AJ, Senis YA, Nash GB, et al. Signalling through Src family kinase isoforms is not redundant in models of thrombo-inflammatory vascular disease. J Cell Mol Med. (2018) 22:4317–27. 10.1111/jcmm.13721
    1. Watson SP, Herbert JM, Pollitt AY. GPVI and CLEC-2 in hemostasis and vascular integrity. J Thromb Haemost. (2010) 8:1456–67. 10.1111/j.1538-7836.2010.03875.x
    1. Li Z, Delaney MK, O'Brien KA, Du X. Signaling during platelet adhesion and activation. Arterioscler Thromb Vasc Biol. (2010) 30:2341–9. 10.1161/ATVBAHA.110.207522
    1. Li Z, Zhang G, Liu J, Stojanovic A, Ruan C, Lowell CA, et al. . An important role of the SRC family kinase Lyn in stimulating platelet granule secretion. J Biol Chem. (2010) 285:12559–70. 10.1074/jbc.M109.098756
    1. Nicolson PLR, Hughes CE, Watson S, Nock SH, Hardy AT, Watson CN, et al. . Inhibition of Btk by Btk-specific concentrations of ibrutinib and acalabrutinib delays but does not block platelet aggregation to GPVI. Haematologica. (2018) 103:2097–108. 10.3324/haematol.2018.193391
    1. Andonegui G, Kerfoot SM, McNagny K, Ebbert KV, Patel KD, Kubes P. Platelets express functional Toll-like receptor-4. Blood. (2005) 106:2417–23. 10.1182/blood-2005-03-0916
    1. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, et al. . Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. (2007) 13:463–9. 10.1038/nm1565
    1. McDonald B, Urrutia R, Yipp BG, Jenne CN, Kubes P. Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis. Cell Host Microbe. (2012) 12:324–33. 10.1016/j.chom.2012.06.011
    1. McDonald B, Davis RP, Kim SJ, Tse M, Esmon CT, Kolaczkowska E, et al. . Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood. (2017) 129:1357–67. 10.1182/blood-2016-09-741298
    1. Martinod K, Fuchs TA, Zitomersky NL, Wong SL, Demers M, Gallant M, et al. PAD4-deficiency does not affect bacteremia in polymicrobial sepsis and ameliorates endotoxemic shock. Blood. (2015) 125:1948–56. 10.1182/blood-2014-07-587709
    1. Jimenez-Alcazar M, Rangaswamy C, Panda R, Bitterling J, Simsek YJ, Long AT, et al. . Host DNases prevent vascular occlusion by neutrophil extracellular traps. Science. (2017) 358:1202–6. 10.1126/science.aam8897
    1. Mai SH, Khan M, Dwivedi DJ, Ross CA, Zhou J, Gould TJ, et al. Delayed but not early treatment with DNase reduces organ damage and improves outcome in a murine model of sepsis. Shock. (2015) 44:166–72. 10.1097/SHK.0000000000000396
    1. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. . Neutrophil extracellular traps kill bacteria. Science. (2004) 303:1532–5. 10.1126/science.1092385

Source: PubMed

3
Abonnere