Yonsei Med J.  2008 Feb;49(1):1-18.

Role of Brain Inflammation in Epileptogenesis

Affiliations
  • 1Department of Pediatrics, Division of Neurology, Northwestern University Children's Memorial Hospital, Chicago, IL, USA. skoh@childrens memorial.org
  • 2Department of Pediatrics, Seoul National University Boramae Hospital, Seoul, Korea.

Abstract

Inflammation is known to participate in the mediation of a growing number of acute and chronic neurological disorders. Even so, the involvement of inflammation in the pathogenesis of epilepsy and seizure-induced brain damage has only recently been appreciated. Inflammatory processes, including activation of microglia and astrocytes and production of proinflammatory cytokines and related molecules, have been described in human epilepsy patients as well as in experimental models of epilepsy. For many decades, a functional role for brain inflammation has been implied by the effective use of anti-inflammatory treatments, such as steroids, in treating intractable pediatric epilepsy of diverse causes. Conversely, common pediatric infectious or autoimmune diseases are often accompanied by seizures during the course of illness. In addition, genetic susceptibility to inflammation correlated with an increased risk of epilepsy. Mounting evidence thus supports the hypothesis that inflammation may contribute to epileptogenesis and cause neuronal injury in epilepsy. We provide an overview of the current knowledge that implicates brain inflammation as a common predisposing factor in epilepsy, particularly childhood epilepsy.

Keyword

Epilepsy; childhood; steroid; cytokine; innate immunity; microglia; astrocytes

MeSH Terms

Animals
Blood-Brain Barrier
Chronic Disease
Encephalitis/genetics/immunology/metabolism/*pathology
Epilepsy/immunology/metabolism/*pathology/therapy
Gene Expression Regulation
Humans
Nervous System Diseases/immunology/pathology

Figure

  • Fig. 1 Acute activation of microglia after KA-induced seizures in CX3cr1EGFP/+ transgenic mice. (A) Low magnification view of a brain section from a Cx3cr1GFP/+ transgenic mouse showing that all parenchymal microglia are fluorescently labeled green because the Cx3cr1-encoding fractalkine chemokine receptor has been replaced by a green fluorescent protein (GFP) reporter gene. The box points to massively accumulated microglia one week after KA-induced seizures, likely at the area of injury, shown in high magnification view (B). High magnification view of PBS control (C) and KA-induced seizures (D). Within two days after status epilepticus, a nearly two-fold increase in the area of fluorescent cells and significant microglial proliferation were noted in KA-treated mice compared to control littermates. Note the ramified processes and apparent increase in the number of GFP+ microglia.

  • Fig. 2 Time course of changes in expression of select inflammation-related genes significantly regulated in rats after KA at P15 and P30 compared to controls. Genes (C) for transcription factors known to be early mediators of immune and inflammation responses, as well as cytokines associated with microglial activation, are rapidly and dramatically up-regulated after KA-induced seizures at P15 (postnatal day 15; juvenile rats) (A) and P30 (postnatal day 30; adult rats) (B). Note that some of these inflammation-related genes are increased to a greater extent than at P15 and are often persistently elevated beyond 72 hours only in adult rats (P30, B) that show seizure-induced cell death and develop spontaneous recurrent seizures.

  • Fig. 3 Triple immunofluorescence confocal images of neurons, microglia, and astrocytes in the epileptogenic cortex of patients with chronic intractable childhood epilepsy. (A) Activated microglia (blue) and astrocytes (red) are found in close contact with each other and with neurons (green). Bar = 100µm. (B) Microglial processes (red) wrap around a neuron (green), engulfing fragmented DNA (blue). Bar = 10µm.


Reference

1. Hauser WA. The prevalence and incidence of convulsive disorders in children. Epilepsia. 1994. 35:Suppl 2. S1–S6.
Article
2. Shinnar S, Pellock JM. Update on the epidemiology and prognosis of pediatric epilepsy. J Child Neurol. 2002. 17:Suppl 1. S4–S17.
Article
3. Berg AT, Shinnar S, Levy SR, Testa FM. Newly diagnosed epilepsy in children: presentation at diagnosis. Epilepsia. 1999. 40:445–452.
Article
4. Hauser WA HD. Epilepsy: Frequency, Causes and Consequences. 1990. New York: Demos.
5. Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004. 16:1–13.
6. Neuhaus J, Risau W, Wolburg H. Induction of blood-brain barrier characteristics in bovine brain endothelial cells by rat astroglial cells in transfilter coculture. Ann N Y Acad Sci. 1991. 633:578–580.
Article
7. Krizanac-Bengez L, Mayberg MR, Janigro D. The cerebral vasculature as a therapeutic target for neurological disorders and the role of shear stress in vascular homeostatis and pathophysiology. Neurol Res. 2004. 26:846–853.
Article
8. Feuerstein GZ, Liu T, Barone FC. Cytokines, inflammation, and brain injury: role of tumor necrosis factor-alpha. Cerebrovasc Brain Metab Rev. 1994. 6:341–360.
9. Tarkowski E, Rosengren L, Blomstrand C, Wikkelsö C, Jensen C, Ekholm S, et al. Intrathecal release of pro- and anti-inflammatory cytokines during stroke. Clin Exp Immunol. 1997. 110:492–499.
Article
10. Rider LG, Thapa PB, Del Beccaro MA, Gale JL, Foy HM, Farwell JR, et al. Cerebrospinal fluid analysis in children with seizures. Pediatr Emerg Care. 1995. 11:226–229.
Article
11. Zhang W, Smith C, Howlett C, Stanimirovic D. Inflammatory activation of human brain endothelial cells by hypoxic astrocytes in vitro is mediated by IL-1beta. J Cereb Blood Flow Metab. 2000. 20:967–978.
Article
12. deVeber G, Roach ES, Riela AR, Wiznitzer M. Stroke in children: recognition, treatment, and future directions. Semin Pediatr Neurol. 2000. 7:309–317.
Article
13. Lynch JK, Nelson KB. Epidemiology of perinatal stroke. Curr Opin Pediatr. 2001. 13:499–505.
Article
14. Kirkham FJ, DeBaun MR. Stroke in Children with Sickle Cell Disease. Curr Treat Options Neurol. 2004. 6:357–375.
Article
15. Aso K, Scher MS, Barmada MA. Cerebral infarcts and seizures in the neonate. J Child Neurol. 1990. 5:224–228.
Article
16. Nelson KB, Lynch JK. Stroke in newborn infants. Lancet Neurol. 2004. 3:150–158.
Article
17. Sreenan C, Bhargava R, Robertson CM. Cerebral infarction in the term newborn: clinical presentation and long-term outcome. J Pediatr. 2000. 137:351–355.
Article
18. Golomb MR, Garg BP, Carvalho KS, Johnson CS, Williams LS. Perinatal stroke and the risk of developing childhood epilepsy. J Pediatr. 2007. 151:409–413. 413.e1–413.e2.
Article
19. Yang JS, Park YD, Hartlage PL. Seizures associated with stroke in childhood. Pediatr Neurol. 1995. 12:136–138.
Article
20. Lanska MJ, Lanska DJ, Horwitz SJ, Aram DM. Presentation, clinical course, and outcome of childhood stroke. Pediatr Neurol. 1991. 7:333–341.
Article
21. Tarkowski E, Rosengren L, Blomstrand C, Wikkelsö C, Jensen C, Ekholm S, et al. Early intrathecal production of interleukin-6 predicts the size of brain lesion in stroke. Stroke. 1995. 26:1393–1398.
Article
22. Cutler SM, Pettus EH, Hoffman SW, Stein DG. Tapered progesterone withdrawal enhances behavioral and molecular recovery after traumatic brain injury. Exp Neurol. 2005. 195:423–429.
Article
23. Lee T, von Scheven E, Sandborg C. Systemic lupus erythematosus and antiphospholipid syndrome in children and adolescents. Curr Opin Rheumatol. 2001. 13:415–421.
Article
24. Toubi E, Khamashta MA, Panarra A, Hughes GR. Association of antiphospholipid antibodies with central nervous system disease in systemic lupus erythematosus. Am J Med. 1995. 99:397–401.
Article
25. Liou HH, Wang CR, Chen CJ, Chen RC, Chuang CY, Chiang IP, et al. Elevated levels of anticardiolipin antibodies and epilepsy in lupus patients. Lupus. 1996. 5:307–312.
Article
26. Watemberg N, Greenstein D, Levine A. Encephalopathy associated with Hashimoto thyroiditis: pediatric perspective. J Child Neurol. 2006. 21:1–5.
Article
27. Stratigos AJ, Laskaris G, Stratigos JD. Behçet's disease. Semin Neurol. 1992. 12:346–357.
Article
28. Joseph FG, Scolding NJ. Neuro-Behçet's disease in Caucasians: a study of 22 patients. Eur J Neurol. 2007. 14:174–180.
Article
29. Mantegazza R, Bernasconi P, Baggi F, Spreafico R, Ragona F, Antozzi C, et al. Antibodies against GluR3 peptides are not specific for Rasmussen's encephalitis but are also present in epilepsy patients with severe, early onset disease and intractable seizures. J Neuroimmunol. 2002. 131:179–185.
Article
30. Peltola JT, Haapala A, Isojärvi JI, Auvinen A, Palmio J, Latvala K, et al. Antiphospholipid and antinuclear antibodies in patients with epilepsy or new-onset seizure disorders. Am J Med. 2000. 109:712–717.
Article
31. Eriksson K, Peltola J, Keränen T, Haapala AM, Koivikko M. High prevalence of antiphospholipid antibodies in children with epilepsy: a controlled study of 50 cases. Epilepsy Res. 2001. 46:129–137.
Article
32. Yoshimura K, Konishi T, Kotani H, Wakiguchi H, Kurashige T. Prevalence of positive anticardiolipin antibody in benign infantile convulsion. Brain Dev. 2001. 23:317–320.
Article
33. Shrivastava A, Dwivedi S, Aggarwal A, Misra R. Anti-cardiolipin and anti-beta2 glycoprotein I antibodies in Indian patients with systemic lupus erythematosus: association with the presence of seizures. Lupus. 2001. 10:45–50.
Article
34. Nelson KB, Ellenberg JH. Predictors of epilepsy in children who have experienced febrile seizures. N Engl J Med. 1976. 295:1029–1033.
Article
35. Millichap JG. Studies in febrile seizures. I. Height of body temperature as a measure of the febrile-seizure threshold. Pediatrics. 1959. 23(1 Pt 1):76–85.
36. Virta M, Hurme M, Helminen M. Increased frequency of interleukin-1beta (-511) allele 2 in febrile seizures. Pediatr Neurol. 2002. 26:192–195.
37. Kanemoto K, Kawasaki J, Yuasa S, Kumaki T, Tomohiro O, Kaji R, et al. Increased frequency of interleukin-1beta-511T allele in patients with temporal lobe epilepsy, hippocampal sclerosis, and prolonged febrile convulsion. Epilepsia. 2003. 44:796–799.
Article
38. Dubé C, Vezzani A, Behrens M, Bartfai T, Baram TZ. Interleukin-1beta contributes to the generation of experimental febrile seizures. Ann Neurol. 2005. 57:152–155.
Article
39. Hall CB, Long CE, Schnabel KC, Caserta MT, McIntyre KM, Costanzo MA, et al. Human herpesvirus-6 infection in children. A prospective study of complications and reactivation. N Engl J Med. 1994. 331:432–438.
Article
40. Chiu SS, Tse CY, Lau YL, Peiris M. Influenza A infection is an important cause of febrile seizures. Pediatrics. 2001. 108:E63.
Article
41. Asano Y, Yoshikawa T, Suga S, Kobayashi I, Nakashima T, Yazaki T, et al. Clinical features of infants with primary human herpesvirus 6 infection (exanthem subitum, roseola infantum). Pediatrics. 1994. 93:104–108.
Article
42. Jee SH, Long CE, Schnabel KC, Sehgal N, Epstein LG, Hall CB. Risk of recurrent seizures after a primary human herpesvirus 6-induced febrile seizure. Pediatr Infect Dis J. 1998. 17:43–48.
Article
43. Bertolani MF, Portolani M, Marotti F, Sabbattini AM, Chiossi C, Bandieri MR, et al. A study of childhood febrile convulsions with particular reference to HHV-6 infection: pathogenic considerations. Childs Nerv Syst. 1996. 12:534–539.
Article
44. Kondo K, Nagafuji H, Hata A, Tomomori C, Yamanishi K. Association of human herpesvirus 6 infection of the central nervous system with recurrence of febrile convulsions. J Infect Dis. 1993. 167:1197–1200.
Article
45. Kawada J, Kimura H, Ito Y, Hara S, Iriyama M, Yoshikawa T, et al. Systemic cytokine responses in patients with influenza-associated encephalopathy. J Infect Dis. 2003. 188:690–698.
Article
46. Masuyama T, Matsuo M, Ichimaru T, Ishii K, Tsuchiya K, Hamasaki Y. Possible contribution of interferon-alpha to febrile seizures in influenza. Pediatr Neurol. 2002. 27:289–292.
47. Vestergaard M, Hviid A, Madsen KM, Wohlfahrt J, Thorsen P, Schendel D, et al. MMR vaccination and febrile seizures: evaluation of susceptible subgroups and long-term prognosis. JAMA. 2004. 292:351–357.
48. Barlow WE, Davis RL, Glasser JW, Rhodes PH, Thompson RS, Mullooly JP, et al. The risk of seizures after receipt of whole-cell pertussis or measles, mumps, and rubella vaccine. N Engl J Med. 2001. 345:656–661.
Article
49. Hirtz DG, Nelson KB, Ellenberg JH. Seizures following childhood immunizations. J Pediatr. 1983. 102:14–18.
Article
50. Contino MF, Lebby T, Arcinue EL. Rotaviral gastrointestinal infection causing afebrile seizures in infancy and childhood. Am J Emerg Med. 1994. 12:94–95.
Article
51. Nishimura S, Ushijima H, Nishimura S, Shiraishi H, Kanazawa C, Abe T, et al. Detection of rotavirus in cerebrospinal fluid and blood of patients with convulsions and gastroenteritis by means of the reverse transcription polymerase chain reaction. Brain Dev. 1993. 15:457–459.
Article
52. Ushijima H, Bosu K, Abe T, Shinozaki T. Suspected rotavirus encephalitis. Arch Dis Child. 1986. 61:692–694.
Article
53. Saulsbury FT, Winkelstein JA, Yolken RH. Chronic rotavirus infection in immunodeficiency. J Pediatr. 1980. 97:61–65.
Article
54. Chen HJ, Chen BS, Wang SF, Lai MH. Rotavirus gastroenteritis in children: a clinical study of 125 patients in Hsin-Tien area. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi. 1991. 32:73–78.
55. Prasad AN, Stafstrom CF, Holmes GL. Alternative epilepsy therapies: the ketogenic diet, immunoglobulins, and steroids. Epilepsia. 1996. 37:Suppl 1. S81–S95.
Article
56. Klein R, Livingston S. The effect of adrenocorticotropic hormone in epilepsy. J Pediatr. 1950. 37:733–742.
Article
57. Hrachovy RA, Frost JD Jr, Glaze DG. High-dose, long-duration versus low-dose, short-duration corticotropin therapy for infantile spasms. The Journal of pediatrics. 1994. 124(5 Pt 1):803–806.
Article
58. Joëls M. Corticosteroid actions in the hippocampus. J Neuroendocrinol. 2001. 13:657–669.
Article
59. Reddy DS, Rogawski MA. Enhanced anticonvulsant activity of ganaxolone after neurosteroid withdrawal in a rat model of catamenial epilepsy. J Pharmacol Exp Ther. 2000. 294:909–915.
60. Baram TZ, Hatalski CG. Neuropeptide-mediated excitability: a key triggering mechanism for seizure generation in the developing brain. Trends Neurosci. 1998. 21:471–476.
Article
61. Rasmussen T, Olszewski J, Lloydsmith D. Focal seizures due to chronic localized encephalitis. Neurology. 1958. 8:435–445.
Article
62. Bauer J, Elger CE, Hans VH, Schramm J, Urbach H, Lassmann H, et al. Astrocytes are a specific immunological target in Rasmussen's encephalitis. Ann Neurol. 2007. 62:67–80.
Article
63. Rogers SW, Andrews PI, Gahring LC, Whisenand T, Cauley K, Crain B, et al. Autoantibodies to glutamate receptor GluR3 in Rasmussen's encephalitis. Science. 1994. 265:648–651.
Article
64. Bahi-Buisson N, Villanueva V, Bulteau C, Delalande O, Dulac O, Chiron C, et al. Long term response to steroid therapy in Rasmussen encephalitis. Seizure. 2007. 16:485–492.
Article
65. Cowan LD, Hudson LS. The epidemiology and natural history of infantile spasms. J Child Neurol. 1991. 6:355–364.
Article
66. Cowan LD, Bodensteiner JB, Leviton A, Doherty L. Prevalence of the epilepsies in children and adolescents. Epilepsia. 1989. 30:94–106.
Article
67. Aicardi J. Aicardi J, editor. Infantile spasms and related syndromes. Epilepsy in children. 1986. New York: Raven Press.
68. Riikonen R. Infantile spasms: some new theoretical aspects. Epilepsia. 1983. 24:159–168.
Article
69. Baram TZ. Pathophysiology of massive infantile spasms: perspective on the putative role of the brain adrenal axis. Ann Neurol. 1993. 33:231–236.
Article
70. Low NL. Infantile spasms with mental retardation. II. Treatment with cortisone and adrenocorticotropin. Pediatrics. 1958. 22:1165–1169.
71. Mackay MT, Weiss SK, Adams-Webber T, Ashwal S, Stephens D, Ballaban-Gill K, et al. Practice parameter: medical treatment of infantile spasms: report of the American Academy of Neurology and the Child Neurology Society. Neurology. 2004. 62:1668–1681.
Article
72. Baram TZ, Mitchell WG, Tournay A, Snead OC, Hanson RA, Horton EJ. High-dose corticotropin (ACTH) versus prednisone for infantile spasms: a prospective, randomized, blinded study. Pediatrics. 1996. 97:375–379.
Article
73. Hrachovy RA, Frost JD Jr, Kellaway P, Zion TE. Double-blind study of ACTH vs prednisone therapy in infantile spasms. J Pediatr. 1983. 103:641–645.
Article
74. Lombroso CT. A prospective study of infantile spasms: clinical and therapeutic correlations. Epilepsia. 1983. 24:135–158.
Article
75. Eisermann MM, DeLaRaillère A, Dellatolas G, Tozzi E, Nabbout R, Dulac O, et al. Infantile spasms in Down syndrome--effects of delayed anticonvulsive treatment. Epilepsy Res. 2003. 55:21–27.
Article
76. Heiskala H. Community-based study of Lennox-Gastaut syndrome. Epilepsia. 1997. 38:526–531.
Article
77. Sinclair DB. Prednisone therapy in pediatric epilepsy. Pediatr Neurol. 2003. 28:194–198.
Article
78. Yamatogi Y, Ohtsuka Y, Ishida T, Ichiba N, Ishida S, Miyake S, et al. Treatment of the Lennox syndrome with ACTH: a clinical and electroencephalographic study. Brain Dev. 1979. 1:267–276.
Article
79. Landau WM, Kleffner FR. Syndrome of acquired aphasia with convulsive disorder in children. Neurology. 1957. 7:523–530.
Article
80. Sinclair DB, Snyder TJ. Corticosteroids for the treatment of Landau-kleffner syndrome and continuous spike-wave discharge during sleep. Pediatr Neurol. 2005. 32:300–306.
Article
81. Tsuru T, Mori M, Mizuguchi M, Momoi MY. Effects of high-dose intravenous corticosteroid therapy in Landau-Kleffner syndrome. Pediatr Neurol. 2000. 22:145–147.
Article
82. Tassinari CA BM, Dravet C, Dalla Bernardina B, Roger J. Roger J, Bureau M, Dravet C, Genton P, Tassinari CA, Wolf P, editors. Epilepsy with continuous spikes and waves during slow sleep-otherwise described as epilepsy with electrical status epilepticus during slow sleep (ESES). Epileptic syndromes in infancy, childhood and adolescence. 2002. 3rd ed. London: John Libbey.
83. Gallagher S, Weiss S, Oram Cardy J, Humphries T, Harman KE, Menascu S. Efficacy of very high dose steroid treatment in a case of Landau-Kleffner syndrome. Dev Med Child Neurol. 2006. 48:766–769.
Article
84. Snead OC 3rd, Benton JW, Myers GJ. ACTH and prednisone in childhood seizure disorders. Neurology. 1983. 33:966–970.
Article
85. Oguni H, Hayashi K, Awaya Y, Fukuyama Y, Osawa M. Severe myoclonic epilepsy in infants--a review based on the Tokyo Women's Medical University series of 84 cases. Brain Dev. 2001. 23:736–748.
Article
86. Verhelst H, Boon P, Buyse G, Ceulemans B, D'Hooghe M, Meirleir LD, et al. Steroids in intractable childhood epilepsy: clinical experience and review of the literature. Seizure. 2005. 14:412–421.
Article
87. Plata-Salamán CR, Ilyin SE, Turrin NP, Gayle D, Flynn MC, Romanovitch AE, et al. Kindling modulates the IL-1beta system, TNF-alpha, TGF-beta1, and neuropeptide mRNAs in specific brain regions. Brain Res Mol Brain Res. 2000. 75:248–258.
Article
88. Zimmer LA, Ennis M, Shipley MT. Soman-induced seizures rapidly activate astrocytes and microglia in discrete brain regions. J Comp Neurol. 1997. 378:482–492.
Article
89. Turrin NP, Rivest S. Innate immune reaction in response to seizures: implications for the neuropathology associated with epilepsy. Neurobiol Dis. 2004. 16:321–334.
Article
90. Jankowsky JL, Patterson PH. The role of cytokines and growth factors in seizures and their sequelae. Prog Neurobiol. 2001. 63:125–149.
Article
91. McNamara JO. Cellular and molecular basis of epilepsy. J Neurosci. 1994. 14:3413–3425.
Article
92. Parent JM, Yu TW, Leibowitz RT, Geschwind DH, Sloviter RS, Lowenstein DH. Dentate granule cell neurogenesis is increased by seizures and contributes to aberrant network reorganization in the adult rat hippocampus. J Neurosci. 1997. 17:3727–3738.
Article
93. Koh S, Storey TW, Santos TC, Mian AY, Cole AJ. Early-life seizures in rats increase susceptibility to seizure-induced brain injury in adulthood. Neurology. 1999. 53:915–921.
Article
94. Aloisi F. Immune function of microglia. Glia. 2001. 36:165–179.
Article
95. Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996. 19:312–318.
Article
96. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005. 76:77–98.
Article
97. Taniwaki Y, Kato M, Araki T, Kobayashi T. Microglial activation by epileptic activities through the propagation pathway of kainic acid-induced hippocampal seizures in the rat. Neurosci Lett. 1996. 217:29–32.
Article
98. Rizzi M, Perego C, Aliprandi M, Richichi C, Ravizza T, Colella D, et al. Glia activation and cytokine increase in rat hippocampus by kainic acid-induced status epilepticus during postnatal development. Neurobiol Dis. 2003. 14:494–503.
Article
99. Ravizza T, Rizzi M, Perego C, Richichi C, Velísková J, Moshé SL, et al. Inflammatory response and glia activation in developing rat hippocampus after status epilepticus. Epilepsia. 2005. 46:Suppl 5. 113–117.
Article
100. Fountain NB. Status epilepticus: risk factors and complications. Epilepsia. 2000. 41:Suppl 2. S23–S30.
Article
101. Henshall DC. Apoptosis signalling pathways in seizure-induced neuronal death and epilepsy. Biochem Soc Trans. 2007. 35(Pt 2):421–423.
Article
102. Kang TC, Kim DS, Kwak SE, Kim JE, Won MH, Kim DW, et al. Epileptogenic roles of astroglial death and regeneration in the dentate gyrus of experimental temporal lobe epilepsy. Glia. 2006. 54:258–271.
Article
103. Streit WJ, Walter SA, Pennell NA. Reactive microgliosis. Prog Neurobiol. 1999. 57:563–581.
Article
104. Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003. 100:13632–13637.
Article
105. Borges K, Gearing M, McDermott DL, Smith AB, Almonte AG, Wainer BH, et al. Neuronal and glial pathological changes during epileptogenesis in the mouse pilocarpine model. Exp Neurol. 2003. 182:21–34.
Article
106. Brandt C, Potschka H, Löscher W, Ebert U. N-methyl-D-aspartate receptor blockade after status epilepticus protects against limbic brain damage but not against epilepsy in the kainate model of temporal lobe epilepsy. Neuroscience. 2003. 118:727–740.
Article
107. Hanisch UK. Microglia as a source and target of cytokines. Glia. 2002. 40:140–155.
Article
108. Aronica E, Gorter JA, Rozemuller AJ, Yankaya B, Troost D. Interleukin-1 beta down-regulates the expression of metabotropic glutamate receptor 5 in cultured human astrocytes. J Neuroimmunol. 2005. 160:188–194.
Article
109. Tilleux S, Berger J, Hermans E. Induction of astrogliosis by activated microglia is associated with a down-regulation of metabotropic glutamate receptor 5. J Neuroimmunol. 2007. 189:23–30.
Article
110. Seifert G, Schilling K, Steinhäuser C. Astrocyte dysfunction in neurological disorders: a molecular perspective. Nat Rev Neurosci. 2006. 7:194–206.
Article
111. Gloor P. Epilepsy surgery. 1999. New York: Raven Press.
112. Tian GF, Azmi H, Takano T, Xu Q, Peng W, Lin J, et al. An astrocytic basis of epilepsy. Nat Med. 2005. 11:973–981.
Article
113. O'Connor ER, Sontheimer H, Spencer DD, de Lanerolle NC. Astrocytes from human hippocampal epileptogenic foci exhibit action potential-like responses. Epilepsia. 1998. 39:347–354.
114. Jørgensen MB, Finsen BR, Jensen MB, Castellano B, Diemer NH, Zimmer J. Microglial and astroglial reactions to ischemic and kainic acid-induced lesions of the adult rat hippocampus. Exp Neurol. 1993. 120:70–88.
Article
115. Somera-Molina KC, Robin B, Somera CA, Anderson C, Stine C, Koh S, et al. Glial activation links early-life seizures and long-term neurologic dysfunction: evidence using a small molecule inhibitor of proinflammatory cytokine upregulation. Epilepsia. 2007. 48:1785–1800.
Article
116. Seifert G, Schröder W, Hinterkeuser S, Schumacher T, Schramm J, Steinhäuser C. Changes in flip/flop splicing of astroglial AMPA receptors in human temporal lobe epilepsy. Epilepsia. 2002. 43:Suppl 5. 162–167.
Article
117. Schröder W, Hinterkeuser S, Seifert G, Schramm J, Jabs R, Wilkin GP, et al. Functional and molecular properties of human astrocytes in acute hippocampal slices obtained from patients with temporal lobe epilepsy. Epilepsia. 2000. 41:Suppl 6. S181–S184.
Article
118. Fedele DE, Gouder N, Guttinger M, Gabernet L, Scheurer L, Rülicke T, et al. Astrogliosis in epilepsy leads to overexpression of adenosine kinase, resulting in seizure aggravation. Brain. 2005. 128(Pt 10):2383–2395.
Article
119. Chung HKS. Encyclopedia of Basic Epilepsy Research. Elsevier: Burlington;in press.
120. Vezzani A, Conti M, De Luigi A, Ravizza T, Moneta D, Marchesi F, et al. Interleukin-1beta immunoreactivity and microglia are enhanced in the rat hippocampus by focal kainate application: functional evidence for enhancement of electrographic seizures. J Neurosci. 1999. 19:5054–5065.
Article
121. Oprica M, Eriksson C, Schultzberg M. Inflammatory mechanisms associated with brain damage induced by kainic acid with special reference to the interleukin-1 system. J Cell Mol Med. 2003. 7:127–140.
Article
122. Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, et al. Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. Proc Natl Acad Sci U S A. 2000. 97:11534–11539.
Article
123. Bernardino L, Xapelli S, Silva AP, Jakobsen B, Poulsen FR, Oliveira CR, et al. Modulator effects of interleukin-1beta and tumor necrosis factor-alpha on AMPA-induced excitotoxicity in mouse organotypic hippocampal slice cultures. J Neurosci. 2005. 25:6734–6744.
Article
124. Ravizza T, Gagliardi B, Noé F, Boer K, Aronica E, Vezzani A. Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimental models and human temporal lobe epilepsy. Neurobiol Dis. 2008. 29:142–160.
Article
125. Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, Von Zastrow M, et al. Control of synaptic strength by glial TNFalpha. Science. 2002. 295:2282–2285.
126. Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T, et al. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci. 2003. 23:8692–8700.
Article
127. Ye ZC, Sontheimer H. Cytokine modulation of glial glutamate uptake: a possible involvement of nitric oxide. Neuroreport. 1996. 7:2181–2185.
128. Bezzi P, Volterra A. A neuron-glia signalling network in the active brain. Curr Opin Neurobiol. 2001. 11:387–394.
Article
129. Samland H, Huitron-Resendiz S, Masliah E, Criado J, Henriksen SJ, Campbell IL. Profound increase in sensitivity to glutamatergic- but not cholinergic agonist-induced seizures in transgenic mice with astrocyte production of IL-6. J Neurosci Res. 2003. 73:176–187.
Article
130. Patel HC, Boutin H, Allan SM. Interleukin-1 in the brain: mechanisms of action in acute neurodegeneration. Ann N Y Acad Sci. 2003. 992:39–47.
131. Rothwell NJ, Luheshi GN. Interleukin 1 in the brain: biology, pathology and therapeutic target. Trends Neurosci. 2000. 23:618–625.
Article
132. Panegyres PK, Hughes J. The neuroprotective effects of the recombinant interleukin-1 receptor antagonist rhIL-1ra after excitotoxic stimulation with kainic acid and its relationship to the amyloid precursor protein gene. J Neurol Sci. 1998. 154:123–132.
Article
133. Giulian D, Li J, Li X, George J, Rutecki PA. The impact of microglia-derived cytokines upon gliosis in the CNS. Dev Neurosci. 1994. 16:128–136.
Article
134. Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter MK, et al. Altered neuronal and microglial responses to excitotoxic and ischemic brain injury in mice lacking TNF receptors. Nat Med. 1996. 2:788–794.
Article
135. Ridet JL, Malhotra SK, Privat A, Gage FH. Reactive astrocytes: cellular and molecular cues to biological function. Trends Neurosci. 1997. 20:570–577.
Article
136. Brett FM, Mizisin AP, Powell HC, Campbell IL. Evolution of neuropathologic abnormalities associated with blood-brain barrier breakdown in transgenic mice expressing interleukin-6 in astrocytes. J Neuropathol Exp Neurol. 1995. 54:766–775.
Article
137. Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB, et al. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci U S A. 1993. 90:10061–10065.
Article
138. Campbell IL. Transgenic mice and cytokine actions in the brain: bridging the gap between structural and functional neuropathology. Brain Res Brain Res Rev. 1998. 26:327–336.
Article
139. Steffensen SC, Campbell IL, Henriksen SJ. Site-specific hippocampal pathophysiology due to cerebral overexpression of interleukin-6 in transgenic mice. Brain Res. 1994. 652:149–153.
Article
140. Penkowa M, Molinero A, Carrasco J, Hidalgo J. Interleukin-6 deficiency reduces the brain inflammatory response and increases oxidative stress and neurodegeneration after kainic acid-induced seizures. Neuroscience. 2001. 102:805–818.
Article
141. Brigande JV, Wieraszko A, Albert MD, Balkema GW, Seyfried TN. Biochemical correlates of epilepsy in the E1 mouse: analysis of glial fibrillary acidic protein and gangliosides. J Neurochem. 1992. 58:752–760.
Article
142. Drage MG, Holmes GL, Seyfried TN. Hippocampal neurons and glia in epileptic EL mice. J Neurocytol. 2002. 31:681–692.
143. Ingram EM, Wiseman JW, Tessler S, Emson PC. Reduction of glial glutamate transporters in the parietal cortex and hippocampus of the EL mouse. J Neurochem. 2001. 79:564–575.
Article
144. Sheng JG, Boop FA, Mrak RE, Griffin WS. Increased neuronal beta-amyloid precursor protein expression in human temporal lobe epilepsy: association with interleukin-1 alpha immunoreactivity. J Neurochem. 1994. 63:1872–1879.
Article
145. Crespel A, Coubes P, Rousset MC, Brana C, Rougier A, Rondouin G, et al. Inflammatory reactions in human medial temporal lobe epilepsy with hippocampal sclerosis. Brain Res. 2002. 952:159–169.
Article
146. Maldonado M, Baybis M, Newman D, Kolson DL, Chen W, McKhann G 2nd, et al. Expression of ICAM-1, TNF-alpha, NF kappa B, and MAP kinase in tubers of the tuberous sclerosis complex. Neurobiol Dis. 2003. 14:279–290.
Article
147. Baranzini SE, Laxer K, Bollen A, Oksenberg JR. Gene expression analysis reveals altered brain transcription of glutamate receptors and inflammatory genes in a patient with chronic focal (Rasmussen's) encephalitis. J Neuroimmunol. 2002. 128:9–15.
Article
148. Vezzani A, Granata T. Brain inflammation in epilepsy: experimental and clinical evidence. Epilepsia. 2005. 46:1724–1743.
Article
149. Hulkkonen J, Koskikallio E, Rainesalo S, Keränen T, Hurme M, Peltola J. The balance of inhibitory and excitatory cytokines is differently regulated in vivo and in vitro among therapy resistant epilepsy patients. Epilepsy Res. 2004. 59:199–205.
Article
150. Peltola J, Hurme M, Miettinen A, Keränen T. Elevated levels of interleukin-6 may occur in cerebrospinal fluid from patients with recent epileptic seizures. Epilepsy Res. 1998. 31:129–133.
Article
151. Peltola J, Palmio J, Korhonen L, Suhonen J, Miettinen A, Hurme M, et al. Interleukin-6 and interleukin-1 receptor antagonist in cerebrospinal fluid from patients with recent tonic-clonic seizures. Epilepsy Res. 2000. 41:205–211.
Article
152. Kira R, Torisu H, Takemoto M, Nomura A, Sakai Y, Sanefuji M, et al. Genetic susceptibility to simple febrile seizures: interleukin-1beta promoter polymorphisms are associated with sporadic cases. Neurosci Lett. 2005. 384:239–244.
Article
153. Beach TG, Woodhurst WB, MacDonald DB, Jones MW. Reactive microglia in hippocampal sclerosis associated with human temporal lobe epilepsy. Neurosci Lett. 1995. 191:27–30.
Article
154. Aronica E, Gorter JA, Redeker S, Ramkema M, Spliet WG, van Rijen PC, et al. Distribution, characterization and clinical significance of microglia in glioneuronal tumours from patients with chronic intractable epilepsy. Neuropathol Appl Neurobiol. 2005. 31:280–291.
Article
155. Boer K, Spliet WG, van Rijen PC, Redeker S, Troost D, Aronica E. Evidence of activated microglia in focal cortical dysplasia. J Neuroimmunol. 2006. 173:188–195.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr