Semin Respir Crit Care Med 2006; 27(4): 365-376
DOI: 10.1055/s-2006-948290
Copyright © 2006 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA.

The Role of the Coagulation Cascade in the Continuum of Sepsis and Acute Lung Injury and Acute Respiratory Distress Syndrome

Julie A. Bastarache1 , Lorraine B. Ware1 , Gordon R. Bernard1
  • 1Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
Further Information

Publication History

Publication Date:
14 August 2006 (online)

ABSTRACT

Sepsis is a common and life-threatening condition with a high mortality rate. Severe sepsis includes multiorgan dysfunction syndrome. The organ most often affected is the lung, with development of acute lung injury (ALI), which, in its most severe form, is referred to as acute respiratory distress syndrome (ARDS). Our understanding of inflammation in the pathogenesis of sepsis and ALI is continually growing. However, therapies aimed at the inflammatory cascade in sepsis have been unsuccessful. These failures have led investigators to consider other pathways that may be important in the development of sepsis and ALI, including the coagulation and fibrinolytic cascades. In fact, the first therapy to reduce mortality in sepsis modulates the coagulation cascade. With this clinical success, administration of drotecogin alfa (recombinant activated protein C), the importance of coagulation in the pathogenesis of human sepsis is becoming clearer. This review summarizes the current understanding of the role of coagulation and fibrinolytic abnormalities in sepsis and the development of ALI and ARDS. Both in vitro and in vivo studies of the role of the coagulation cascade in sepsis and lung injury will be discussed, including initiation of coagulation through modulation of tissue factor and tissue factor pathway inhibitor, propagation of coagulation via protein C and thrombomodulin, inhibition of thrombin generation and resolution through thrombolysis by plasminogen activator, and plasminogen activator inhibitor-1.

REFERENCES

  • 1 Martin G S, Mannino D M, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000.  N Engl J Med. 2003;  348 1546-1554
  • 2 Bochud P Y, Calandra T. Pathogenesis of sepsis: new concepts and implications for future treatment.  BMJ. 2003;  326 262-266
  • 3 Hudson L D, Milberg J A, Anardi D, Maunder R J. Clinical risks for development of the acute respiratory distress syndrome.  Am J Respir Crit Care Med. 1995;  151(2 Pt 1) 293-301
  • 4 Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network.  N Engl J Med. 2000;  342 1301-1308
  • 5 American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis.  Crit Care Med. 1992;  20 864-874
  • 6 Bernard G R, Vincent J L, Laterre P F et al.. Efficacy and safety of recombinant human activated protein C for severe sepsis.  N Engl J Med. 2001;  344 699-709
  • 7 Bernard G R, Artigas A, Brigham K L et al.. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination.  Am J Respir Crit Care Med. 1994;  149(3 Pt 1) 818-824
  • 8 Dixon B. The role of microvascular thrombosis in sepsis.  Anaesth Intensive Care. 2004;  32 619-629
  • 9 Bachofen M, Weibel E R. Structural alterations of lung parenchyma in the adult respiratory distress syndrome.  Clin Chest Med. 1982;  3 35-56
  • 10 Steinhoff M, Buddenkotte J, Shpacovitch V et al.. Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response.  Endocr Rev. 2005;  26 1-43
  • 11 Esmon C T. The impact of the inflammatory response on coagulation.  Thromb Res. 2004;  114 321-327
  • 12 Bernard G R, Luce J M, Sprung C L et al.. High-dose corticosteroids in patients with the adult respiratory distress syndrome.  N Engl J Med. 1987;  317 1565-1570
  • 13 Luce J M, Montgomery A B, Marks J D, Turner J, Metz C A, Murray J F. Ineffectiveness of high-dose methylprednisolone in preventing parenchymal lung injury and improving mortality in patients with septic shock.  Am Rev Respir Dis. 1988;  138 62-68
  • 14 Meduri G U, Headley A S, Golden E et al.. Effect of prolonged methylprednisolone therapy in unresolving acute respiratory distress syndrome: a randomized controlled trial.  JAMA. 1998;  280 159-165
  • 15 Ketoconazole for early treatment of acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. The ARDS Network.  JAMA. 2000;  283 1995-2002
  • 16 Moons A H, Levi M, Peters R J. Tissue factor and coronary artery disease.  Cardiovasc Res. 2002;  53 313-325
  • 17 Belting M, Dorrell M I, Sandgren S et al.. Regulation of angiogenesis by tissue factor cytoplasmic domain signaling.  Nat Med. 2004;  10 502-509
  • 18 Voigtlander C, Rand A, Liu S L et al.. Suppression of tissue factor expression, cofactor activity, and metastatic potential of murine melanoma cells by the N-terminal domain of adenovirus E1A 12S protein.  J Cell Biochem. 2002;  85 54-71
  • 19 Wolberg A S, Kon R H, Monroe D M, Ezban M, Roberts H R, Hoffman M. Deencryption of cellular tissue factor is independent of its cytoplasmic domain.  Biochem Biophys Res Commun. 2000;  272 332-336
  • 20 McVey J H. Tissue factor pathway.  Baillieres Best Pract Res Clin Haematol. 1999;  12 361-372
  • 21 Price G C, Thompson S A, Kam P C. Tissue factor and tissue factor pathway inhibitor.  Anaesthesia. 2004;  59 483-492
  • 22 Mattsson E, Hartung T, Morath S, Egesten A. Highly purified lipoteichoic acid from Staphylococcus aureus induces procoagulant activity and tissue factor expression in human monocytes but is a weak inducer in whole blood: comparison with peptidoglycan.  Infect Immun. 2004;  72 4322-4326
  • 23 Guha M, O'Connell M A, Pawlinski R et al.. Lipopolysaccharide activation of the MEK-ERK1/2 pathway in human monocytic cells mediates tissue factor and tumor necrosis factor alpha expression by inducing Elk-1 phosphorylation and Egr-1 expression.  Blood. 2001;  98 1429-1439
  • 24 Henriksson C E, Klingenberg O, Ovstebo R, Joo G B, Westvik A B, Kierulf P. Discrepancy between tissue factor activity and tissue factor expression in endotoxin-induced monocytes is associated with apoptosis and necrosis.  Thromb Haemost. 2005;  94 1236-1244
  • 25 Egorina E M, Sovershaev M A, Bjorkoy G et al.. Intracellular and surface distribution of monocyte tissue factor: application to intersubject variability.  Arterioscler Thromb Vasc Biol. 2005;  25 1493-1498
  • 26 Egorina E M, Sovershaev M A, Osterud B. In-Cell Western assay: a new approach to visualize tissue factor in human monocytes.  J Thromb Haemost. 2006;  4 614-620
  • 27 Lupu C, Westmuckett A D, Peer G et al.. Tissue factor-dependent coagulation is preferentially up-regulated within arterial branching areas in a baboon model of Escherichia coli sepsis.  Am J Pathol. 2005;  167 1161-1172
  • 28 Erlich J, Fearns C, Mathison J, Ulevitch R J, Mackman N. Lipopolysaccharide induction of tissue factor expression in rabbits.  Infect Immun. 1999;  67 2540-2546
  • 29 Dackiw A P, McGilvray I D, Woodside M, Nathens A B, Marshall J C, Rotstein O D. Prevention of endotoxin-induced mortality by antitissue factor immunization.  Arch Surg. 1996;  131 1273-1278 discussion 8-9
  • 30 Taylor Jr F B, Chang A, Ruf W et al.. Lethal E. coli septic shock is prevented by blocking tissue factor with monoclonal antibody.  Circ Shock. 1991;  33 127-134
  • 31 Levi M, ten Cate H, Bauer K A et al.. Inhibition of endotoxin-induced activation of coagulation and fibrinolysis by pentoxifylline or by a monoclonal anti-tissue factor antibody in chimpanzees.  J Clin Invest. 1994;  93 114-120
  • 32 Warr T A, Rao L V, Rapaport S I. Disseminated intravascular coagulation in rabbits induced by administration of endotoxin or tissue factor: effect of anti-tissue factor antibodies and measurement of plasma extrinsic pathway inhibitor activity.  Blood. 1990;  75 1481-1489
  • 33 Marsik C, Quehenberger P, Mackman N, Osterud B, Luther T, Jilma B. Validation of a novel tissue factor assay in experimental human endotoxemia.  Thromb Res. 2003;  111 311-315
  • 34 Marsik C, Mayr F, Cardona F, Derhaschnig U, Wagner O F, Jilma B. Endotoxaemia modulates Toll-like receptors on leucocytes in humans.  Br J Haematol. 2003;  121 653-656
  • 35 de Jonge E, Dekkers P E, Creasey A A et al.. Tissue factor pathway inhibitor dose-dependently inhibits coagulation activation without influencing the fibrinolytic and cytokine response during human endotoxemia.  Blood. 2000;  95 1124-1129
  • 36 Jilma B, Marsik C, Mayr F et al.. Pharmacodynamics of active site-inhibited factor VIIa in endotoxin-induced coagulation in humans.  Clin Pharmacol Ther. 2002;  72 403-410
  • 37 Hogan K A, Weiler H, Lord S T. Mouse models in coagulation.  Thromb Haemost. 2002;  87 563-574
  • 38 Pawlinski R, Fernandes A, Kehrle B et al.. Tissue factor deficiency causes cardiac fibrosis and left ventricular dysfunction.  Proc Natl Acad Sci U S A. 2002;  99 15333-15338
  • 39 Pawlinski R, Pedersen B, Schabbauer G et al.. Role of tissue factor and protease-activated receptors in a mouse model of endotoxemia.  Blood. 2004;  103 1342-1347
  • 40 Carraway M S, Welty-Wolf K E, Miller D L et al.. Blockade of tissue factor: treatment for organ injury in established sepsis.  Am J Respir Crit Care Med. 2003;  167 1200-1209
  • 41 Welty-Wolf K E, Carraway M S, Ortel T L et al.. Blockade of tissue factor-factor X binding attenuates sepsis-induced respiratory and renal failure.  Am J Physiol Lung Cell Mol Physiol. 2006;  290(1) L21-L31
  • 42 Nakstad B, Boye N P, Lyberg T. Procoagulant activities in human alveolar macrophages.  Eur J Respir Dis. 1987;  71 459-471
  • 43 Tomashefski Jr J F, Davies P, Boggis C, Greene R, Zapol W M, Reid L M. The pulmonary vascular lesions of the adult respiratory distress syndrome.  Am J Pathol. 1983;  112 112-126
  • 44 Lang I M, Mackman N, Kriett J M, Moser K M, Schleef R R. Prothrombotic activation of pulmonary arterial endothelial cells in a patient with tuberculosis.  Hum Pathol. 1996;  27 423-427
  • 45 Marshall B C, Brown B R, Rothstein M A, Rao N V, Hoidal J R, Rodgers G M. Alveolar epithelial cells express both plasminogen activator and tissue factor: potential role in repair of lung injury.  Chest. 1991;  99(Suppl 3) 25S-27S
  • 46 Gross T J, Simon R H, Sitrin R G. Tissue factor procoagulant expression by rat alveolar epithelial cells.  Am J Respir Cell Mol Biol. 1992;  6 397-403
  • 47 Drake T A, Morrissey J H, Edgington T S. Selective cellular expression of tissue factor in human tissues: implications for disorders of hemostasis and thrombosis.  Am J Pathol. 1989;  134 1087-1097
  • 48 Prudhomme J B, Geiser T, Matthay M A, Ware L B. The alveolar epithelium can initiate the extrinsic coagulation cascade through expression of tissue factor on the cell surface [abstract].  Am J Respir Crit Care Med. 2004;  169 A414
  • 49 Jesmin S, Gando S, Matsuda N et al.. Temporal changes in pulmonary expression of key procoagulant molecules in rabbits with endotoxin-induced acute lung injury: elevated expression levels of protease-activated receptors.  Thromb Haemost. 2004;  92 966-979
  • 50 Armstead V E, Opentanova I L, Minchenko A G, Lefer A M. Tissue factor expression in vital organs during murine traumatic shock: role of transcription factors AP-1 and NF-kappaB.  Anesthesiology. 1999;  91 1844-1852
  • 51 Olman M A, Mackman N, Gladson C L, Moser K M, Loskutoff D J. Changes in procoagulant and fibrinolytic gene expression during bleomycin-induced lung injury in the mouse.  J Clin Invest. 1995;  96 1621-1630
  • 52 Idell S, Peters J, James K K, Fair D S, Coalson J J. Local abnormalities of coagulation and fibrinolytic pathways that promote alveolar fibrin deposition in the lungs of baboons with diffuse alveolar damage.  J Clin Invest. 1989;  84 181-193
  • 53 Enkhbaatar P, Okajima K, Murakami K et al.. Recombinant tissue factor pathway inhibitor reduces lipopolysaccharide-induced pulmonary vascular injury by inhibiting leukocyte activation.  Am J Respir Crit Care Med. 2000;  162 1752-1759
  • 54 Miller D L, Welty-Wolf K, Carraway M S et al.. Extrinsic coagulation blockade attenuates lung injury and proinflammatory cytokine release after intratracheal lipopolysaccharide.  Am J Respir Cell Mol Biol. 2002;  26 650-658
  • 55 Idell S, Koenig K B, Fair D S, Martin T R, McLarty J, Maunder R J. Serial abnormalities of fibrin turnover in evolving adult respiratory distress syndrome.  Am J Physiol. 1991;  261(4 Pt 1) L240-L248
  • 56 Fuchs-Buder T, de Moerloose P, Ricou B et al.. Time course of procoagulant activity and D dimer in bronchoalveolar fluid of patients at risk for or with acute respiratory distress syndrome.  Am J Respir Crit Care Med. 1996;  153 163-167
  • 57 Gunther A, Mosavi P, Heinemann S et al.. Alveolar fibrin formation caused by enhanced procoagulant and depressed fibrinolytic capacities in severe pneumonia: comparison with the acute respiratory distress syndrome.  Am J Respir Crit Care Med. 2000;  161(2 Pt 1) 454-462
  • 58 Gando S, Nanzaki S, Morimoto Y, Kobayashi S, Kemmotsu O. Systemic activation of tissue-factor dependent coagulation pathway in evolving acute respiratory distress syndrome in patients with trauma and sepsis.  J Trauma. 1999;  47 719-723
  • 59 Abraham E, Reinhart K, Opal S et al.. Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled trial.  JAMA. 2003;  290 238-247
  • 60 Esmon C T, Stenflo J, Suttie J W. A new vitamin K-dependent protein: a phospholipid-binding zymogen of a serine esterase.  J Biol Chem. 1976;  251 3052-3056
  • 61 Esmon C T. The anticoagulant and anti-inflammatory roles of the protein C anticoagulant pathway.  J Autoimmun. 2000;  15 113-116
  • 62 Esmon C T. The protein C pathway.  Chest. 2003;  124(Suppl 3) 26S-32S
  • 63 Stearns-Kurosawa D J, Kurosawa S, Mollica J S, Ferrell G L, Esmon C T. The endothelial cell protein C receptor augments protein C activation by the thrombin-thrombomodulin complex.  Proc Natl Acad Sci U S A. 1996;  93 10212-10216
  • 64 Taylor Jr F B, Chang A, Esmon C T, D'Angelo A, Vigano-D'Angelo S, Blick K E. Protein C prevents the coagulopathic and lethal effects of Escherichia coli infusion in the baboon.  J Clin Invest. 1987;  79 918-925
  • 65 Powars D R, Rogers Z R, Patch M J, McGehee W G, Francis Jr R B. Purpura fulminans in meningococcemia: association with acquired deficiencies of proteins C and S.  N Engl J Med. 1987;  317 571-572
  • 66 Mannucci P M, Vigano S. Deficiencies of protein C, an inhibitor of blood coagulation.  Lancet. 1982;  2(8296) 463-467
  • 67 Vigano-D' Angelo S, Comp P C, Esmon C T, D'Angelo A. Relationship between protein C antigen and anticoagulant activity during oral anticoagulation and in selected disease states.  J Clin Invest. 1986;  77 416-425
  • 68 Grey S T, Tsuchida A, Hau H, Orthner C L, Salem H H, Hancock W W. Selective inhibitory effects of the anticoagulant activated protein C on the responses of human mononuclear phagocytes to LPS, IFN-gamma, or phorbol ester.  J Immunol. 1994;  153 3664-3672
  • 69 White B, Schmidt M, Murphy C et al.. Activated protein C inhibits lipopolysaccharide-induced nuclear translocation of nuclear factor kappaB (NF-kappaB) and tumour necrosis factor alpha (TNF-alpha) production in the THP-1 monocytic cell line.  Br J Haematol. 2000;  110 130-134
  • 70 Hirose K, Okajima K, Taoka Y et al.. Activated protein C reduces the ischemia/reperfusion-induced spinal cord injury in rats by inhibiting neutrophil activation.  Ann Surg. 2000;  232 272-280
  • 71 Sturn D H, Kaneider N C, Feistritzer C, Djanani A, Fukudome K, Wiedermann C J. Expression and function of the endothelial protein C receptor in human neutrophils.  Blood. 2003;  102 1499-1505
  • 72 Derhaschnig U, Reiter R, Knobl P, Baumgartner M, Keen P, Jilma B. Recombinant human activated protein C (rhAPC; drotrecogin alfa [activated]) has minimal effect on markers of coagulation, fibrinolysis, and inflammation in acute human endotoxemia.  Blood. 2003;  102 2093-2098
  • 73 Kalil A C, Coyle S M, Um J Y et al.. Effects of drotrecogin alfa (activated) in human endotoxemia.  Shock. 2004;  21 222-229
  • 74 Idell S. Endothelium and disordered fibrin turnover in the injured lung: newly recognized pathways.  Crit Care Med. 2002;  30(Suppl 5) S274-S280
  • 75 Cheng T, Liu D, Griffin J H et al.. Activated protein C blocks p53-mediated apoptosis in ischemic human brain endothelium and is neuroprotective.  Nat Med. 2003;  9 338-342
  • 76 Esmon C T. Protein C pathway in sepsis.  Ann Med. 2002;  34 598-605
  • 77 Murakami K, Okajima K, Uchiba M et al.. Activated protein C prevents LPS-induced pulmonary vascular injury by inhibiting cytokine production.  Am J Physiol. 1997;  272(2 Pt 1) L197-L202
  • 78 Redl H, Schlag G, Schiesser A, Davies J. Thrombomodulin release in baboon sepsis: its dependence on the dose of Escherichia coli and the presence of tumor necrosis factor.  J Infect Dis. 1995;  171 1522-1527
  • 79 Gu J M, Katsuura Y, Ferrell G L, Grammas P, Esmon C T. Endotoxin and thrombin elevate rodent endothelial cell protein C receptor mRNA levels and increase receptor shedding in vivo.  Blood. 2000;  95 1687-1693
  • 80 Taylor Jr F B, Stearns-Kurosawa D J, Kurosawa S et al.. The endothelial cell protein C receptor aids in host defense against Escherichia coli sepsis.  Blood. 2000;  95 1680-1686
  • 81 Yan S B, Helterbrand J D, Hartman D L, Wright T J, Bernard G R. Low levels of protein C are associated with poor outcome in severe sepsis.  Chest. 2001;  120 915-922
  • 82 Liaw P C, Esmon C T, Kahnamoui K et al.. Patients with severe sepsis vary markedly in their ability to generate activated protein C.  Blood. 2004;  104 3958-3964
  • 83 Ware L B, Fang X, Matthay M A. Protein C and thrombomodulin in human acute lung injury.  Am J Physiol Lung Cell Mol Physiol. 2003;  285 L514-L521
  • 84 Dhainaut J F, Laterre P F, Janes J M et al.. Drotrecogin alfa (activated) in the treatment of severe sepsis patients with multiple-organ dysfunction: data from the PROWESS trial.  Intensive Care Med. 2003;  29 894-903
  • 85 Nick J A, Coldren C D, Geraci M W et al.. Recombinant human activated protein C reduces human endotoxin-induced pulmonary inflammation via inhibition of neutrophil chemotaxis.  Blood. 2004;  104 3878-3885
  • 86 Mondino A, Blasi F. uPA and uPAR in fibrinolysis, immunity and pathology.  Trends Immunol. 2004;  25 450-455
  • 87 Yepes M, Lawrence D A. New functions for an old enzyme: nonhemostatic roles for tissue-type plasminogen activator in the central nervous system.  Exp Biol Med (Maywood). 2004;  229 1097-1104
  • 88 Fay W P. Plasminogen activator inhibitor 1, fibrin, and the vascular response to injury.  Trends Cardiovasc Med. 2004;  14 196-202
  • 89 Albers G W, Amarenco P, Easton J D, Sacco R L, Teal P. Antithrombotic and thrombolytic therapy for ischemic stroke: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy.  Chest. 2004;  126(Suppl 3) 483S-512S
  • 90 Dunn C J, Goa K L. Tenecteplase: a review of its pharmacology and therapeutic efficacy in patients with acute myocardial infarction.  Am J Cardiovasc Drugs. 2001;  1 51-66
  • 91 Goldhaber S Z, Bounameaux H. Thrombolytic therapy in pulmonary embolism.  Semin Vasc Med. 2001;  1 213-220
  • 92 Romer J, Nielsen B S, Ploug M. The urokinase receptor as a potential target in cancer therapy.  Curr Pharm Des. 2004;  10 2359-2376
  • 93 Chorostowska-Wynimko J, Skrzypczak-Jankun E, Jankun J. Plasminogen activator inhibitor type-1: its structure, biological activity and role in tumorigenesis (Review).  Int J Mol Med. 2004;  13 759-766
  • 94 Alessi M C, Juhan-Vague I. Contribution of PAI-1 in cardiovascular pathology.  Arch Mal Coeur Vaiss. 2004;  97 673-678
  • 95 Texereau J, Pene F, Chiche J D, Rousseau C, Mira J P. Importance of hemostatic gene polymorphisms for susceptibility to and outcome of severe sepsis.  Crit Care Med. 2004;  32(Suppl 5) S313-S319
  • 96 Biemond B J, Levi M, Ten Cate H et al.. Plasminogen activator and plasminogen activator inhibitor I release during experimental endotoxaemia in chimpanzees: effect of interventions in the cytokine and coagulation cascades.  Clin Sci (Lond). 1995;  88 587-594
  • 97 Zeerleder S, Schroeder V, Hack C E, Kohler H P, Wuillemin W A. TAFI and PAI-1 levels in human sepsis.  Thromb Res. 2006;  118(2) 205-212
  • 98 Eriksson P, Kallin B, van't Hooft F M, Bavenholm P, Hamsten A. Allele-specific increase in basal transcription of the plasminogen-activator inhibitor 1 gene is associated with myocardial infarction.  Proc Natl Acad Sci USA. 1995;  92 1851-1855
  • 99 Hermans P W, Hazelzet J A. Plasminogen activator inhibitor type 1 gene polymorphism and sepsis.  Clin Infect Dis. 2005;  41(Suppl 7) S453-S458
  • 100 Westendorp R G, Hottenga J J, Slagboom P E. Variation in plasminogen-activator-inhibitor-1 gene and risk of meningococcal septic shock.  Lancet. 1999;  354 561-563
  • 101 Haralambous E, Hibberd M L, Hermans P W, Ninis N, Nadel S, Levin M. Role of functional plasminogen-activator-inhibitor-1 4G/5G promoter polymorphism in susceptibility, severity, and outcome of meningococcal disease in Caucasian children.  Crit Care Med. 2003;  31 2788-2793
  • 102 Mesters R M, Florke N, Ostermann H, Kienast J. Increase of plasminogen activator inhibitor levels predicts outcome of leukocytopenic patients with sepsis.  Thromb Haemost. 1996;  75 902-907
  • 103 Hou B, Eren M, Painter C A et al.. Tumor necrosis factor alpha activates the human plasminogen activator inhibitor-1 gene through a distal nuclear factor kappaB site.  J Biol Chem. 2004;  279 18127-18136
  • 104 Montes R, Rodriguez-Whilhelmi P, Hurtado V et al.. The endotoxin-induced plasminogen activator inhibitor-1 increase in rabbits is not tumor necrosis factor-alpha dependent and can occur in the absence of interleukin-1beta.  Thromb Haemost. 2002;  88 639-643
  • 105 Saetre T, Lindgaard A K, Lyberg T. Systemic activation of coagulation and fibrinolysis in a porcine model of serogroup A streptococcal shock.  Blood Coagul Fibrinolysis. 2000;  11 433-438
  • 106 Pechlaner C, Buratti T, Biedermann H, Joannidis M, Wiedermann C J. Thrombolytic therapy in adult meningococcal purpura fulminans with acute renal failure and severe perfusion deficits to the extremities.  Wien Klin Wochenschr. 1999;  111 650-654
  • 107 Aiuto L T, Barone S R, Cohen P S, Boxer R A. Recombinant tissue plasminogen activator restores perfusion in meningococcal purpura fulminans.  Crit Care Med. 1997;  25 1079-1082
  • 108 Chapman Jr H A, Stone O L, Vavrin Z. Degradation of fibrin and elastin by intact human alveolar macrophages in vitro: characterization of a plasminogen activator and its role in matrix degradation.  J Clin Invest. 1984;  73 806-815
  • 109 Chapman Jr H A, Stone O L. A fibrinolytic inhibitor of human alveolar macrophages: induction with endotoxin.  Am Rev Respir Dis. 1985;  132 569-575
  • 110 Chapman H A, Yang X L, Sailor L Z, Sugarbaker D J. Developmental expression of plasminogen activator inhibitor type 1 by human alveolar macrophages: possible role in lung injury.  J Immunol. 1990;  145 3398-3405
  • 111 Wygrecka M, Markart P, Ruppert C et al.. Compartment- and cell-specific expression of coagulation and fibrinolysis factors in the murine lung undergoing inhalational versus intravenous endotoxin application.  Thromb Haemost. 2004;  92 529-540
  • 112 Ts'ao C, Ward W F. Plasminogen activator activity in lung and alveolar macrophages of rats exposed to graded single doses of gamma rays to the right hemithorax.  Radiat Res. 1985;  103 393-402
  • 113 Carley W W, Niedbala M J, Gerritsen M E. Isolation, cultivation, and partial characterization of microvascular endothelium derived from human lung.  Am J Respir Cell Mol Biol. 1992;  7 620-630
  • 114 Grau G E, de Moerloose P, Bulla O et al.. Haemostatic properties of human pulmonary and cerebral microvascular endothelial cells.  Thromb Haemost. 1997;  77 585-590
  • 115 Prabhakaran P, Ware L B, White K E, Cross M T, Matthay M A, Olman M A. Elevated levels of plasminogen activator inhibitor-1 in pulmonary edema fluid are associated with mortality in acute lung injury.  Am J Physiol Lung Cell Mol Physiol. 2003;  285 L20-L28
  • 116 Moalli R, Doyle J M, Tahhan H R, Hasan F M, Braman S S, Saldeen T. Fibrinolysis in critically ill patients.  Am Rev Respir Dis. 1989;  140 287-293
  • 117 Groeneveld A B, Kindt I, Raijmakers P G, Hack C E, Thijs L G. Systemic coagulation and fibrinolysis in patients with or at risk for the adult respiratory distress syndrome.  Thromb Haemost. 1997;  78 1444-1449
  • 118 Ware L B, Eisner M D, Wickersham N et al.. Plasminogen activator inhibitor-1 (PAI-1), a marker of impaired fibrinolysis, is associated with higher mortality in patients with ALI/ARDS [abstract]. Am J Respir Crit Care Med 2004 169: A115
  • 119 Baudo F, Caimi T M, de Cataldo F et al.. Antithrombin III (ATIII) replacement therapy in patients with sepsis and/or postsurgical complications: a controlled double-blind, randomized, multicenter study.  Intensive Care Med. 1998;  24 336-342
  • 120 Inthorn D, Hoffmann J N, Hartl W H, Muhlbayer D, Jochum M. Antithrombin III supplementation in severe sepsis: beneficial effects on organ dysfunction.  Shock. 1997;  8 328-334
  • 121 Fourrier F, Chopin C, Huart J J, Runge I, Caron C, Goudemand J. Double-blind, placebo-controlled trial of antithrombin III concentrates in septic shock with disseminated intravascular coagulation.  Chest. 1993;  104 882-888
  • 122 Warren B L, Eid A, Singer P et al.. Caring for the critically ill patient: high-dose antithrombin III in severe sepsis: a randomized controlled trial.  JAMA. 2001;  286 1869-1878
  • 123 Wiedermann C J, Hoffmann J N, Juers M et al.. High-dose antithrombin III in the treatment of severe sepsis in patients with a high risk of death: efficacy and safety.  Crit Care Med. 2006;  34 285-292

Julie
A BastaracheM.D. 

T1218 MCN

1161 21st Ave. S, Nashville, TN 37232-2650

Email: julie.bastarache@vanderbilt.edu

    >