Semin Thromb Hemost 2013; 39(08): 876-880
DOI: 10.1055/s-0033-1357482
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Coagulation in Inflammatory Diseases of the Central Nervous System

Joab Chapman
1   Department of Neurology and Joseph Sagol Neuroscience Center, Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Israel
› Author Affiliations
Further Information

Publication History

Publication Date:
09 October 2013 (online)

Abstract

Thrombin and other proteases involved in coagulation also have the potential to stimulate inflammation in the brain to a large extent through the protease-activated receptors (PARs). Such exposure of the brain to increased levels of coagulation factors is more likely to occur during vasculitis and activation of intrinsic coagulation in the brain and may cause inflammatory diseases such as multiple sclerosis. There is growing evidence from animal models and human brain samples that links upregulation of coagulation factors to inflammatory central nervous system (CNS) disease. Evidence includes measures of thrombin-like activity, levels of its receptors, PARs, and inhibitors of the coagulation pathway. The major receptor for thrombin, PAR-1, has now been definitively located to the synapse and node of Ranvier and its excessive activation leads to blocks in long-term potentiation and in nerve conduction. There is growing evidence that modulating coagulation in models of CNS inflammatory disease has beneficial clinical effects. These findings suggest that coagulation-like pathways play a significant role in the pathogenesis of inflammatory diseases in the CNS and present a viable target for therapeutic strategies.

 
  • References

  • 1 Deschepper CF, Bigornia V, Berens ME, Lapointe MC. Production of thrombin and antithrombin III by brain and astroglial cell cultures. Brain Res Mol Brain Res 1991; 11 (3-4) 355-358
  • 2 Dihanich M, Kaser M, Reinhard E, Cunningham D, Monard D. Prothrombin mRNA is expressed by cells of the nervous system. Neuron 1991; 6 (4) 575-581
  • 3 Ferland G. Vitamin K, an emerging nutrient in brain function. Biofactors 2012; 38 (2) 151-157
  • 4 Davalos D, Ryu JK, Merlini M , et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun 2012; 3: 1227
  • 5 Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol 2012; 34 (1) 43-62
  • 6 Ryu JK, Davalos D, Akassoglou K. Fibrinogen signal transduction in the nervous system. J Thromb Haemost 2009; 7 (Suppl. 01) 151-154
  • 7 Mullins ES, Kombrinck KW, Talmage KE , et al. Genetic elimination of prothrombin in adult mice is not compatible with survival and results in spontaneous hemorrhagic events in both heart and brain. Blood 2009; 113 (3) 696-704
  • 8 Bar-Shavit R, Hruska KA, Kahn AJ, Wilner GD. Hormone-like activity of human thrombin. Ann N Y Acad Sci 1986; 485: 335-348
  • 9 Coughlin SR. Thrombin signalling and protease-activated receptors. Nature 2000; 407 (6801) 258-264
  • 10 Weinstein JR, Zhang M, Kutlubaev M , et al. Thrombin-induced regulation of CD95(Fas) expression in the N9 microglial cell line: evidence for involvement of proteinase-activated receptor(1) and extracellular signal-regulated kinase 1/2. Neurochem Res 2009; 34 (3) 445-452
  • 11 Möller T, Weinstein JR, Hanisch UK. Activation of microglial cells by thrombin: past, present, and future. Semin Thromb Hemost 2006; 32 (Suppl. 01) 69-76
  • 12 Hanisch UK, van Rossum D, Xie Y , et al. The microglia-activating potential of thrombin: the protease is not involved in the induction of proinflammatory cytokines and chemokines. J Biol Chem 2004; 279 (50) 51880-51887
  • 13 Möller T, Hanisch UK, Ransom BR. Thrombin-induced activation of cultured rodent microglia. J Neurochem 2000; 75 (4) 1539-1547
  • 14 Gurwitz D, Cunningham DD. Thrombin modulates and reverses neuroblastoma neurite outgrowth. Proc Natl Acad Sci U S A 1988; 85 (10) 3440-3444
  • 15 Cunningham DD, Donovan FM. Regulation of neurons and astrocytes by thrombin and protease nexin-1. Relationship to brain injury. Adv Exp Med Biol 1997; 425: 67-75
  • 16 Schmidlin F, Bunnett NW. Protease-activated receptors: how proteases signal to cells. Curr Opin Pharmacol 2001; 1 (6) 575-582
  • 17 Vu TK, Hung DT, Wheaton VI, Coughlin SR. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell 1991; 64 (6) 1057-1068
  • 18 Déry O, Corvera CU, Steinhoff M, Bunnett NW. Proteinase-activated receptors: novel mechanisms of signaling by serine proteases. Am J Physiol 1998; 274 (6, Pt 1) C1429-C1452
  • 19 Donovan FM, Cunningham DD. Signaling pathways involved in thrombin-induced cell protection. J Biol Chem 1998; 273 (21) 12746-12752
  • 20 Pike CJ, Vaughan PJ, Cunningham DD, Cotman CW. Thrombin attenuates neuronal cell death and modulates astrocyte reactivity induced by beta-amyloid in vitro. J Neurochem 1996; 66 (4) 1374-1382
  • 21 Festoff BW, Smirnova IV, Ma J, Citron BA. Thrombin, its receptor and protease nexin I, its potent serpin, in the nervous system. Semin Thromb Hemost 1996; 22 (3) 267-271
  • 22 Cirino G, Cicala C, Bucci MR, Sorrentino L, Maraganore JM, Stone SR. Thrombin functions as an inflammatory mediator through activation of its receptor. J Exp Med 1996; 183 (3) 821-827
  • 23 Lee KR, Drury I, Vitarbo E, Hoff JT. Seizures induced by intracerebral injection of thrombin: a model of intracerebral hemorrhage. J Neurosurg 1997; 87 (1) 73-78
  • 24 Xue M, Del Bigio MR. Acute tissue damage after injections of thrombin and plasmin into rat striatum. Stroke 2001; 32 (9) 2164-2169
  • 25 Lee KR, Colon GP, Betz AL, Keep RF, Kim S, Hoff JT. Edema from intracerebral hemorrhage: the role of thrombin. J Neurosurg 1996; 84 (1) 91-96
  • 26 Aronovich R, Gurwitz D, Kloog Y, Chapman J. Antiphospholipid antibodies, thrombin and LPS activate brain endothelial cells and Ras-dependent pathways through distinct mechanisms. Immunobiology 2005; 210 (10) 781-788
  • 27 Alabanza LM, Bynoe MS. Thrombin induces an inflammatory phenotype in a human brain endothelial cell line. J Neuroimmunol 2012; 245 (1-2) 48-55
  • 28 Menachem A, Chapman J, Katzav A. Significant changes in the levels of secreted cytokines in brains of experimental antiphospholipid syndrome mice. Autoimmune Dis 2012; 2012: 404815
  • 29 Tanne D, Katzav A, Beilin O , et al. Interaction of inflammation, thrombosis, aspirin and enoxaparin in CNS experimental antiphospholipid syndrome. Neurobiol Dis 2008; 30 (1) 56-64
  • 30 Katzav A, Grigoriadis NC, Ebert T , et al. Coagulopathy triggered autoimmunity: experimental antiphospholipid syndrome in factor V Leiden mice. BMC Med 2013; 11: 92
  • 31 Katzav A, Kivity S, Blank M, Shoenfeld Y, Chapman J. Adjuvant immunization induces high levels of pathogenic antiphospholipid antibodies in genetically prone mice: another facet of the ASIA syndrome. Lupus 2012; 21 (2) 210-216
  • 32 Frauenknecht K, Katzav A, Grimm C, Chapman J, Sommer CJ. Neurological impairment in experimental antiphospholipid syndrome is associated with increased ligand binding to hippocampal and cortical serotonergic 5-HT1A receptors. Immunobiology 2013; 218 (4) 517-526
  • 33 Katzav A, Litvinjuk Y, Pick CG , et al. Genetic and immunological factors interact in a mouse model of CNS antiphospholipid syndrome. Behav Brain Res 2006; 169 (2) 289-293
  • 34 Katzav A, Pick CG, Korczyn AD , et al. Hyperactivity in a mouse model of the antiphospholipid syndrome. Lupus 2001; 10 (7) 496-499
  • 35 Shrot S, Katzav A, Korczyn AD , et al. Behavioral and cognitive deficits occur only after prolonged exposure of mice to antiphospholipid antibodies. Lupus 2002; 11 (11) 736-743
  • 36 Rosidi NL, Zhou J, Pattanaik S , et al. Cortical microhemorrhages cause local inflammation but do not trigger widespread dendrite degeneration. PLoS ONE 2011; 6 (10) e26612
  • 37 Giwa MO, Williams J, Elderfield K , et al. Neuropathologic evidence of endothelial changes in cerebral small vessel disease. Neurology 2012; 78 (3) 167-174
  • 38 Cicala C, Cirino G. Linkage between inflammation and coagulation: an update on the molecular basis of the crosstalk. Life Sci 1998; 62 (20) 1817-1824
  • 39 Esmon CT. Role of coagulation inhibitors in inflammation. Thromb Haemost 2001; 86 (1) 51-56
  • 40 Miller KD, Phelan AW, Collins DN. Prothrombin activation by encephalitogenic brain protein. Exp Mol Pathol 1966; 5 (6) 575-579
  • 41 Koh CS, Gausas J, Paterson PY. Concordance and localization of maximal vascular permeability change and fibrin deposition in the central neuraxis of Lewis rats with cell-transferred experimental allergic encephalomyelitis. J Neuroimmunol 1992; 38 (1–2) 85-93
  • 42 Inaba Y, Ichikawa M, Inoue A , et al. Plasma thrombin-antithrombin III complex is associated with the severity of experimental autoimmune encephalomyelitis. J Neurol Sci 2001; 185 (2) 89-93
  • 43 Beilin O, Gurwitz D, Korczyn AD, Chapman J. Quantitative measurements of mouse brain thrombin-like and thrombin inhibition activities. Neuroreport 2001; 12 (11) 2347-2351
  • 44 Guenther J, Nick H, Monard D. A glia-derived neurite-promoting factor with protease inhibitory activity. EMBO J 1985; 4 (8) 1963-1966
  • 45 Gloor S, Odink K, Guenther J, Nick H, Monard D. A glia-derived neurite promoting factor with protease inhibitory activity belongs to the protease nexins. Cell 1986; 47 (5) 687-693
  • 46 Reinhard E, Suidan HS, Pavlik A, Monard D. Glia-derived nexin/protease nexin-1 is expressed by a subset of neurons in the rat brain. J Neurosci Res 1994; 37 (2) 256-270
  • 47 Wagner SL, Geddes JW, Cotman CW , et al. Protease nexin-1, an antithrombin with neurite outgrowth activity, is reduced in Alzheimer disease. Proc Natl Acad Sci U S A 1989; 86 (21) 8284-8288
  • 48 Kalaria RN, Golde T, Kroon SN, Perry G. Serine protease inhibitor antithrombin III and its messenger RNA in the pathogenesis of Alzheimer's disease. Am J Pathol 1993; 143 (3) 886-893
  • 49 Deschepper CF, Bigornia V, Berens ME, Lapointe MC. Production of thrombin and antithrombin III by brain and astroglial cell cultures. Brain Res Mol Brain Res 1991; 11 (3–4) 355-358
  • 50 Beilin O, Karussis DM, Korczyn AD , et al. Increased thrombin inhibition in experimental autoimmune encephalomyelitis. J Neurosci Res 2005; 79 (3) 351-359
  • 51 Beilin O, Karussis DM, Korczyn AD , et al. Increased KPI containing amyloid precursor protein in experimental autoimmune encephalomyelitis brains. Neuroreport 2007; 18 (6) 581-584
  • 52 Chelmicka-Szorc E, Arnason BG. Partial suppression of experimental allergic encephalomyelitis with heparin. Arch Neurol 1972; 27 (2) 153-158
  • 53 Inaba Y, Ichikawa M, Koh CS , et al. Suppression of experimental autoimmune encephalomyelitis by dermatan sulfate. Cell Immunol 1999; 198 (2) 96-102
  • 54 Wiedermann ChJ, Römisch J. The anti-inflammatory actions of antithrombin—a review. Acta Med Austriaca 2002; 29 (3) 89-92
  • 55 Traugott U, Raine CS, McFarlin DE. Acute experimental allergic encephalomyelitis in the mouse: immunopathology of the developing lesion. Cell Immunol 1985; 91 (1) 240-254
  • 56 Cuzner ML, Opdenakker G. Plasminogen activators and matrix metalloproteases, mediators of extracellular proteolysis in inflammatory demyelination of the central nervous system. J Neuroimmunol 1999; 94 (1–2) 1-14
  • 57 Teesalu T, Hinkkanen AE, Vaheri A. Coordinated induction of extracellular proteolysis systems during experimental autoimmune encephalomyelitis in mice. Am J Pathol 2001; 159 (6) 2227-2237
  • 58 Kieseier BC, Seifert T, Giovannoni G, Hartung HP. Matrix metalloproteinases in inflammatory demyelination: targets for treatment. Neurology 1999; 53 (1) 20-25
  • 59 Suidan HS, Bouvier J, Schaerer E, Stone SR, Monard D, Tschopp J. Granzyme A released upon stimulation of cytotoxic T lymphocytes activates the thrombin receptor on neuronal cells and astrocytes. Proc Natl Acad Sci U S A 1994; 91 (17) 8112-8116
  • 60 Eeckhout Y, Vaes G. Further studies on the activation of procollagenase, the latent precursor of bone collagenase. Effects of lysosomal cathepsin B, plasmin and kallikrein, and spontaneous activation. Biochem J 1977; 166 (1) 21-31
  • 61 Meins M, Piosik P, Schaeren-Wiemers N , et al. Progressive neuronal and motor dysfunction in mice overexpressing the serine protease inhibitor protease nexin-1 in postmitotic neurons. J Neurosci 2001; 21 (22) 8830-8841
  • 62 Houenou LJ, Turner PL, Li L, Oppenheim RW, Festoff BW. A serine protease inhibitor, protease nexin I, rescues motoneurons from naturally occurring and axotomy-induced cell death. Proc Natl Acad Sci U S A 1995; 92 (3) 895-899
  • 63 Marx J. A new link in the brain's defenses. Science 1992; 256 (5061) 1278-1280
  • 64 Campos-de-Magalhães M, de Almeida AJ, Papaiz-Alvarenga RM, Gadelha T, Morais-de-Sá CA, Alves-Leon SV. Normal plasma antithrombin activity in patients with relapsing-remitting and secondary progressive multiple sclerosis. Clin Neurol Neurosurg 2009; 111 (5) 407-411
  • 65 Han MH, Hwang SI, Roy DB , et al. Proteomic analysis of active multiple sclerosis lesions reveals therapeutic targets. Nature 2008; 451 (7182) 1076-1081
  • 66 East E, Gverić D, Baker D, Pryce G, Lijnen HR, Cuzner ML. Chronic relapsing experimental allergic encephalomyelitis (CREAE) in plasminogen activator inhibitor-1 knockout mice: the effect of fibrinolysis during neuroinflammation. Neuropathol Appl Neurobiol 2008; 34 (2) 216-230
  • 67 East E, Baker D, Pryce G, Lijnen HR, Cuzner ML, Gverić D. A role for the plasminogen activator system in inflammation and neurodegeneration in the central nervous system during experimental allergic encephalomyelitis. Am J Pathol 2005; 167 (2) 545-554
  • 68 Shavit E, Beilin O, Korczyn AD , et al. Thrombin receptor PAR-1 on myelin at the node of Ranvier: a new anatomy and physiology of conduction block. Brain 2008; 131 (Pt 4) 1113-1122
  • 69 Shavit E, Michaelson DM, Chapman J. Anatomical localization of protease-activated receptor-1 and protease-mediated neuroglial crosstalk on peri-synaptic astrocytic endfeet. J Neurochem 2011; 119 (3) 460-473
  • 70 Maggio N, Shavit E, Chapman J, Segal M. Thrombin induces long-term potentiation of reactivity to afferent stimulation and facilitates epileptic seizures in rat hippocampal slices: toward understanding the functional consequences of cerebrovascular insults. J Neurosci 2008; 28 (3) 732-736
  • 71 Almonte AG, Qadri LH, Sultan FA , et al. Protease-activated receptor-1 modulates hippocampal memory formation and synaptic plasticity. J Neurochem 2013; 124 (1) 109-122
  • 72 Almonte AG, Sweatt JD. Serine proteases, serine protease inhibitors, and protease-activated receptors: roles in synaptic function and behavior. Brain Res 2011; 1407: 107-122
  • 73 Maggio N, Blatt I, Vlachos A, Tanne D, Chapman J, Segal M. Treating seizures and epilepsy with anticoagulants?. Front Cell Neurosci 2013; 7: 19
  • 74 Maggio N, Itsekson Z, Dominissini D , et al. Thrombin regulation of synaptic plasticity: Implications for physiology and pathology. Exp Neurol 2013; 247: 595-604
  • 75 Maggio N, Cavaliere C, Papa M, Blatt I, Chapman J, Segal M. Thrombin regulation of synaptic transmission: implications for seizure onset. Neurobiol Dis 2013; 50: 171-178