Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2017

Open Access 01-12-2017 | Review

Novel pathomechanisms in inflammatory neuropathies

Authors: David Schafflick, Bernd C. Kieseier, Heinz Wiendl, Gerd Meyer zu Horste

Published in: Journal of Neuroinflammation | Issue 1/2017

Login to get access

Abstract

Inflammatory neuropathies are rare autoimmune-mediated disorders affecting the peripheral nervous system. Considerable progress has recently been made in understanding pathomechanisms of these disorders which will be essential for developing novel diagnostic and therapeutic strategies in the future. Here, we summarize our current understanding of antigenic targets and the relevance of new immunological concepts for inflammatory neuropathies. In addition, we provide an overview of available animal models of acute and chronic variants and how new diagnostic tools such as magnetic resonance imaging and novel therapeutic candidates will benefit patients with inflammatory neuropathies in the future. This review thus illustrates the gap between pre-clinical and clinical findings and aims to outline future directions of development.
Literature
1.
go back to reference van Koningsveld R, Steyerberg EW, Hughes RA, Swan AV, van Doorn PA, Jacobs BC. A clinical prognostic scoring system for Guillain-Barre syndrome. Lancet Neurol. 2007;6:589–94.PubMedCrossRef van Koningsveld R, Steyerberg EW, Hughes RA, Swan AV, van Doorn PA, Jacobs BC. A clinical prognostic scoring system for Guillain-Barre syndrome. Lancet Neurol. 2007;6:589–94.PubMedCrossRef
2.
go back to reference Kuwabara S, Misawa S, Mori M, Tamura N, Kubota M, Hattori T. Long term prognosis of chronic inflammatory demyelinating polyneuropathy: a five year follow up of 38 cases. J Neurol Neurosurg Psychiatry. 2006;77:66–70.PubMedPubMedCentralCrossRef Kuwabara S, Misawa S, Mori M, Tamura N, Kubota M, Hattori T. Long term prognosis of chronic inflammatory demyelinating polyneuropathy: a five year follow up of 38 cases. J Neurol Neurosurg Psychiatry. 2006;77:66–70.PubMedPubMedCentralCrossRef
3.
go back to reference van Doorn PA, Ruts L, Jacobs BC. Clinical features, pathogenesis, and treatment of Guillain-Barre syndrome. Lancet Neurol. 2008;7:939–50.PubMedCrossRef van Doorn PA, Ruts L, Jacobs BC. Clinical features, pathogenesis, and treatment of Guillain-Barre syndrome. Lancet Neurol. 2008;7:939–50.PubMedCrossRef
4.
go back to reference Griffin JW, Sheikh K. The Guillain-Barré Syndromes. In: Dyck PJ, Thomas PK, editors. Peripheral neuropathy, vol. 2. 4th ed. Philadelphia: Elsevier Saunders; 2005. p. 2197–220.CrossRef Griffin JW, Sheikh K. The Guillain-Barré Syndromes. In: Dyck PJ, Thomas PK, editors. Peripheral neuropathy, vol. 2. 4th ed. Philadelphia: Elsevier Saunders; 2005. p. 2197–220.CrossRef
5.
go back to reference Fokke C, van den Berg B, Drenthen J, Walgaard C, van Doorn PA, Jacobs BC. Diagnosis of Guillain-Barre syndrome and validation of Brighton criteria. Brain. 2014;137:33–43.PubMedCrossRef Fokke C, van den Berg B, Drenthen J, Walgaard C, van Doorn PA, Jacobs BC. Diagnosis of Guillain-Barre syndrome and validation of Brighton criteria. Brain. 2014;137:33–43.PubMedCrossRef
6.
go back to reference Sejvar JJ, Baughman AL, Wise M, Morgan OW. Population incidence of Guillain-Barre syndrome: a systematic review and meta-analysis. Neuroepidemiology. 2011;36:123–33.PubMedCrossRefPubMedCentral Sejvar JJ, Baughman AL, Wise M, Morgan OW. Population incidence of Guillain-Barre syndrome: a systematic review and meta-analysis. Neuroepidemiology. 2011;36:123–33.PubMedCrossRefPubMedCentral
7.
go back to reference Walgaard C, Lingsma HF, Ruts L, van Doorn PA, Steyerberg EW, Jacobs BC. Early recognition of poor prognosis in Guillain-Barre syndrome. Neurology. 2011;76:968–75.PubMedPubMedCentralCrossRef Walgaard C, Lingsma HF, Ruts L, van Doorn PA, Steyerberg EW, Jacobs BC. Early recognition of poor prognosis in Guillain-Barre syndrome. Neurology. 2011;76:968–75.PubMedPubMedCentralCrossRef
8.
go back to reference Meyer zu Horste G, Hartung HP, Kieseier BC. From bench to bedside--experimental rationale for immune-specific therapies in the inflamed peripheral nerve. Nat Clin Pract Neurol. 2007;3:198–211.PubMedCrossRef Meyer zu Horste G, Hartung HP, Kieseier BC. From bench to bedside--experimental rationale for immune-specific therapies in the inflamed peripheral nerve. Nat Clin Pract Neurol. 2007;3:198–211.PubMedCrossRef
9.
go back to reference Dalakas MC. Advances in the diagnosis, pathogenesis and treatment of CIDP. Nat Rev Neurol. 2011;7:507–17.PubMedCrossRef Dalakas MC. Advances in the diagnosis, pathogenesis and treatment of CIDP. Nat Rev Neurol. 2011;7:507–17.PubMedCrossRef
10.
go back to reference Vallat JM, Sommer C, Magy L. Chronic inflammatory demyelinating polyradiculoneuropathy: diagnostic and therapeutic challenges for a treatable condition. Lancet Neurol. 2010;9:402–12.PubMedCrossRef Vallat JM, Sommer C, Magy L. Chronic inflammatory demyelinating polyradiculoneuropathy: diagnostic and therapeutic challenges for a treatable condition. Lancet Neurol. 2010;9:402–12.PubMedCrossRef
11.
go back to reference Koller H, Kieseier BC, Jander S, Hartung HP. Chronic inflammatory demyelinating polyneuropathy. N Engl J Med. 2005;352:1343–56.PubMedCrossRef Koller H, Kieseier BC, Jander S, Hartung HP. Chronic inflammatory demyelinating polyneuropathy. N Engl J Med. 2005;352:1343–56.PubMedCrossRef
12.
go back to reference Chio A, Cocito D, Bottacchi E, Buffa C, Leone M, Plano F, Mutani R, Calvo A, Parcidp. Idiopathic chronic inflammatory demyelinating polyneuropathy: an epidemiological study in Italy. J Neurol Neurosurg Psychiatry. 2007;78:1349–53.PubMedPubMedCentralCrossRef Chio A, Cocito D, Bottacchi E, Buffa C, Leone M, Plano F, Mutani R, Calvo A, Parcidp. Idiopathic chronic inflammatory demyelinating polyneuropathy: an epidemiological study in Italy. J Neurol Neurosurg Psychiatry. 2007;78:1349–53.PubMedPubMedCentralCrossRef
13.
go back to reference Iijima M, Koike H, Hattori N, Tamakoshi A, Katsuno M, Tanaka F, Yamamoto M, Arimura K, Sobue G, Refractory Peripheral Neuropathy Study Group of J. Prevalence and incidence rates of chronic inflammatory demyelinating polyneuropathy in the Japanese population. J Neurol Neurosurg Psychiatry. 2008;79:1040–3.PubMedCrossRef Iijima M, Koike H, Hattori N, Tamakoshi A, Katsuno M, Tanaka F, Yamamoto M, Arimura K, Sobue G, Refractory Peripheral Neuropathy Study Group of J. Prevalence and incidence rates of chronic inflammatory demyelinating polyneuropathy in the Japanese population. J Neurol Neurosurg Psychiatry. 2008;79:1040–3.PubMedCrossRef
14.
go back to reference Laughlin RS, Dyck PJ, Melton LJ 3rd, Leibson C, Ransom J, Dyck PJ. Incidence and prevalence of CIDP and the association of diabetes mellitus. Neurology. 2009;73:39–45.PubMedPubMedCentralCrossRef Laughlin RS, Dyck PJ, Melton LJ 3rd, Leibson C, Ransom J, Dyck PJ. Incidence and prevalence of CIDP and the association of diabetes mellitus. Neurology. 2009;73:39–45.PubMedPubMedCentralCrossRef
15.
go back to reference Lunn MP, Manji H, Choudhary PP, Hughes RA, Thomas PK. Chronic inflammatory demyelinating polyradiculoneuropathy: a prevalence study in south east England. J Neurol Neurosurg Psychiatry. 1999;66:677–80.PubMedPubMedCentralCrossRef Lunn MP, Manji H, Choudhary PP, Hughes RA, Thomas PK. Chronic inflammatory demyelinating polyradiculoneuropathy: a prevalence study in south east England. J Neurol Neurosurg Psychiatry. 1999;66:677–80.PubMedPubMedCentralCrossRef
16.
go back to reference Rajabally YA, Simpson BS, Beri S, Bankart J, Gosalakkal JA. Epidemiologic variability of chronic inflammatory demyelinating polyneuropathy with different diagnostic criteria: study of a UK population. Muscle Nerve. 2009;39:432–8.PubMedCrossRef Rajabally YA, Simpson BS, Beri S, Bankart J, Gosalakkal JA. Epidemiologic variability of chronic inflammatory demyelinating polyneuropathy with different diagnostic criteria: study of a UK population. Muscle Nerve. 2009;39:432–8.PubMedCrossRef
17.
go back to reference Mahdi-Rogers M, Hughes RA. Epidemiology of chronic inflammatory neuropathies in southeast England. Eur J Neurol. 2014;21:28–33.PubMedCrossRef Mahdi-Rogers M, Hughes RA. Epidemiology of chronic inflammatory neuropathies in southeast England. Eur J Neurol. 2014;21:28–33.PubMedCrossRef
18.
go back to reference Dyck PJ, Lais AC, Ohta M, Bastron JA, Okazaki H, Groover RV. Chronic inflammatory polyradiculoneuropathy. Mayo Clin Proc. 1975;50:621–37.PubMed Dyck PJ, Lais AC, Ohta M, Bastron JA, Okazaki H, Groover RV. Chronic inflammatory polyradiculoneuropathy. Mayo Clin Proc. 1975;50:621–37.PubMed
20.
21.
go back to reference Sommer C, Koch S, Lammens M, Gabreels-Festen A, Stoll G, Toyka KV. Macrophage clustering as a diagnostic marker in sural nerve biopsies of patients with CIDP. Neurology. 2005;65:1924–9.PubMedCrossRef Sommer C, Koch S, Lammens M, Gabreels-Festen A, Stoll G, Toyka KV. Macrophage clustering as a diagnostic marker in sural nerve biopsies of patients with CIDP. Neurology. 2005;65:1924–9.PubMedCrossRef
22.
go back to reference Vital C, Vital A, Lagueny A, Ferrer X, Fontan D, Barat M, Gbikpi-Benissan G, Orgogozo JM, Henry P, Brechenmacher C, et al. Chronic inflammatory demyelinating polyneuropathy: immunopathological and ultrastructural study of peripheral nerve biopsy in 42 cases. Ultrastruct Pathol. 2000;24:363–9.PubMedCrossRef Vital C, Vital A, Lagueny A, Ferrer X, Fontan D, Barat M, Gbikpi-Benissan G, Orgogozo JM, Henry P, Brechenmacher C, et al. Chronic inflammatory demyelinating polyneuropathy: immunopathological and ultrastructural study of peripheral nerve biopsy in 42 cases. Ultrastruct Pathol. 2000;24:363–9.PubMedCrossRef
23.
go back to reference Hughes RA, Donofrio P, Bril V, Dalakas MC, Deng C, Hanna K, Hartung HP, Latov N, Merkies IS, van Doorn PA. Intravenous immune globulin (10% caprylate-chromatography purified) for the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (ICE study): a randomised placebo-controlled trial. Lancet Neurol. 2008;7:136–44.PubMedCrossRef Hughes RA, Donofrio P, Bril V, Dalakas MC, Deng C, Hanna K, Hartung HP, Latov N, Merkies IS, van Doorn PA. Intravenous immune globulin (10% caprylate-chromatography purified) for the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (ICE study): a randomised placebo-controlled trial. Lancet Neurol. 2008;7:136–44.PubMedCrossRef
24.
go back to reference van den Berg B, Walgaard C, Drenthen J, Fokke C, Jacobs BC, van Doorn PA. Guillain-Barre syndrome: pathogenesis, diagnosis, treatment and prognosis. Nat Rev Neurol. 2014;10:469–82.PubMedCrossRef van den Berg B, Walgaard C, Drenthen J, Fokke C, Jacobs BC, van Doorn PA. Guillain-Barre syndrome: pathogenesis, diagnosis, treatment and prognosis. Nat Rev Neurol. 2014;10:469–82.PubMedCrossRef
25.
go back to reference Mathey EK, Park SB, Hughes RA, Pollard JD, Armati PJ, Barnett MH, Taylor BV, Dyck PJ, Kiernan MC, Lin CS. Chronic inflammatory demyelinating polyradiculoneuropathy: from pathology to phenotype. J Neurol Neurosurg Psychiatry. 2015;86:973–85.PubMedPubMedCentralCrossRef Mathey EK, Park SB, Hughes RA, Pollard JD, Armati PJ, Barnett MH, Taylor BV, Dyck PJ, Kiernan MC, Lin CS. Chronic inflammatory demyelinating polyradiculoneuropathy: from pathology to phenotype. J Neurol Neurosurg Psychiatry. 2015;86:973–85.PubMedPubMedCentralCrossRef
26.
go back to reference Hughes RA, Hadden RD, Gregson NA, Smith KJ. Pathogenesis of Guillain-Barre syndrome. J Neuroimmunol. 1999;100:74–97.PubMedCrossRef Hughes RA, Hadden RD, Gregson NA, Smith KJ. Pathogenesis of Guillain-Barre syndrome. J Neuroimmunol. 1999;100:74–97.PubMedCrossRef
27.
29.
go back to reference Araujo LM, Ferreira ML, Nascimento OJ. Guillain-Barre syndrome associated with the Zika virus outbreak in Brazil. Arq Neuropsiquiatr. 2016;74:253–5.PubMedCrossRef Araujo LM, Ferreira ML, Nascimento OJ. Guillain-Barre syndrome associated with the Zika virus outbreak in Brazil. Arq Neuropsiquiatr. 2016;74:253–5.PubMedCrossRef
30.
go back to reference de Oliveira WK, Carmo EH, Henriques CM, Coelho G, Vazquez E, Cortez-Escalante J, Molina J, Aldighieri S, Espinal MA, Dye C. Zika virus infection and associated neurologic disorders in Brazil. N Engl J Med. 2017;376:1591–3.PubMedPubMedCentralCrossRef de Oliveira WK, Carmo EH, Henriques CM, Coelho G, Vazquez E, Cortez-Escalante J, Molina J, Aldighieri S, Espinal MA, Dye C. Zika virus infection and associated neurologic disorders in Brazil. N Engl J Med. 2017;376:1591–3.PubMedPubMedCentralCrossRef
31.
go back to reference Song BH, Yun SI, Woolley M, Lee YM. Zika virus: history, epidemiology, transmission, and clinical presentation. J Neuroimmunol. 2017;308:50–64.PubMedCrossRef Song BH, Yun SI, Woolley M, Lee YM. Zika virus: history, epidemiology, transmission, and clinical presentation. J Neuroimmunol. 2017;308:50–64.PubMedCrossRef
32.
go back to reference Krauer F, Riesen M, Reveiz L, Oladapo OT, Martinez-Vega R, Porgo TV, Haefliger A, Broutet NJ, Low N, Group WHOZCW. Zika virus infection as a cause of congenital brain abnormalities and Guillain-Barre syndrome: systematic review. PLoS Med. 2017;14:e1002203.PubMedPubMedCentralCrossRef Krauer F, Riesen M, Reveiz L, Oladapo OT, Martinez-Vega R, Porgo TV, Haefliger A, Broutet NJ, Low N, Group WHOZCW. Zika virus infection as a cause of congenital brain abnormalities and Guillain-Barre syndrome: systematic review. PLoS Med. 2017;14:e1002203.PubMedPubMedCentralCrossRef
33.
go back to reference Cao-Lormeau VM, Blake A, Mons S, Lastere S, Roche C, Vanhomwegen J, Dub T, Baudouin L, Teissier A, Larre P, et al. Guillain-Barre syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet. 2016;387:1531–9.PubMedPubMedCentralCrossRef Cao-Lormeau VM, Blake A, Mons S, Lastere S, Roche C, Vanhomwegen J, Dub T, Baudouin L, Teissier A, Larre P, et al. Guillain-Barre syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet. 2016;387:1531–9.PubMedPubMedCentralCrossRef
34.
go back to reference da Silva IRF, Frontera JA, Bispo de Filippis AM, Nascimento O, Group R-G-ZR. Neurologic complications associated with the Zika virus in Brazilian adults. JAMA Neurol. 2017;74:1190–8.PubMedCrossRef da Silva IRF, Frontera JA, Bispo de Filippis AM, Nascimento O, Group R-G-ZR. Neurologic complications associated with the Zika virus in Brazilian adults. JAMA Neurol. 2017;74:1190–8.PubMedCrossRef
35.
go back to reference Parra B, Lizarazo J, Jimenez-Arango JA, Zea-Vera AF, Gonzalez-Manrique G, Vargas J, Angarita JA, Zuniga G, Lopez-Gonzalez R, Beltran CL, et al. Guillain-Barre syndrome associated with Zika virus infection in Colombia. N Engl J Med. 2016;375:1513–23.PubMedCrossRef Parra B, Lizarazo J, Jimenez-Arango JA, Zea-Vera AF, Gonzalez-Manrique G, Vargas J, Angarita JA, Zuniga G, Lopez-Gonzalez R, Beltran CL, et al. Guillain-Barre syndrome associated with Zika virus infection in Colombia. N Engl J Med. 2016;375:1513–23.PubMedCrossRef
36.
go back to reference Lucchese G, Kanduc D. Zika virus and autoimmunity: from microcephaly to Guillain-Barre syndrome, and beyond. Autoimmun Rev. 2016;15:801–8.PubMedCrossRef Lucchese G, Kanduc D. Zika virus and autoimmunity: from microcephaly to Guillain-Barre syndrome, and beyond. Autoimmun Rev. 2016;15:801–8.PubMedCrossRef
37.
go back to reference Ritter JM, Martines RB, Zaki SR. Zika virus: pathology from the pandemic. Arch Pathol Lab Med. 2017;141:49–59.PubMedCrossRef Ritter JM, Martines RB, Zaki SR. Zika virus: pathology from the pandemic. Arch Pathol Lab Med. 2017;141:49–59.PubMedCrossRef
38.
go back to reference Illa I, Ortiz N, Gallard E, Juarez C, Grau JM, Dalakas MC. Acute axonal Guillain-Barre syndrome with IgG antibodies against motor axons following parenteral gangliosides. Ann Neurol. 1995;38:218–24.PubMedCrossRef Illa I, Ortiz N, Gallard E, Juarez C, Grau JM, Dalakas MC. Acute axonal Guillain-Barre syndrome with IgG antibodies against motor axons following parenteral gangliosides. Ann Neurol. 1995;38:218–24.PubMedCrossRef
39.
go back to reference Morikawa M, Kuwahara M, Ueno R, Samukawa M, Hamada Y, Kusunoki S. Serological study using glycoarray for detecting antibodies to glycolipids and glycolipid complexes in immune-mediated neuropathies. J Neuroimmunol. 2016;301:35–40.PubMedCrossRef Morikawa M, Kuwahara M, Ueno R, Samukawa M, Hamada Y, Kusunoki S. Serological study using glycoarray for detecting antibodies to glycolipids and glycolipid complexes in immune-mediated neuropathies. J Neuroimmunol. 2016;301:35–40.PubMedCrossRef
40.
go back to reference Rink C, Gortzen A, Veh RW, Pruss H. Serum antibodies targeting neurons of the monoaminergic systems in Guillain-Barre syndrome. J Neurol Sci. 2017;372:318–23.PubMedCrossRef Rink C, Gortzen A, Veh RW, Pruss H. Serum antibodies targeting neurons of the monoaminergic systems in Guillain-Barre syndrome. J Neurol Sci. 2017;372:318–23.PubMedCrossRef
41.
go back to reference Kusunoki S, Shimizu J, Chiba A, Ugawa Y, Hitoshi S, Kanazawa I. Experimental sensory neuropathy induced by sensitization with ganglioside GD1b. Ann Neurol. 1996;39:424–31.PubMedCrossRef Kusunoki S, Shimizu J, Chiba A, Ugawa Y, Hitoshi S, Kanazawa I. Experimental sensory neuropathy induced by sensitization with ganglioside GD1b. Ann Neurol. 1996;39:424–31.PubMedCrossRef
42.
go back to reference Moyano AL, Comin R, Lardone RD, Alaniz ME, Theaux R, Irazoqui FJ, Nores GA. Validation of a rabbit model of neuropathy induced by immunization with gangliosides. J Neurol Sci. 2008;272:110–4.PubMedCrossRef Moyano AL, Comin R, Lardone RD, Alaniz ME, Theaux R, Irazoqui FJ, Nores GA. Validation of a rabbit model of neuropathy induced by immunization with gangliosides. J Neurol Sci. 2008;272:110–4.PubMedCrossRef
43.
go back to reference Yuki N, Yamada M, Koga M, Odaka M, Susuki K, Tagawa Y, Ueda S, Kasama T, Ohnishi A, Hayashi S, et al. Animal model of axonal Guillain-Barre syndrome induced by sensitization with GM1 ganglioside. Ann Neurol. 2001;49:712–20.PubMedCrossRef Yuki N, Yamada M, Koga M, Odaka M, Susuki K, Tagawa Y, Ueda S, Kasama T, Ohnishi A, Hayashi S, et al. Animal model of axonal Guillain-Barre syndrome induced by sensitization with GM1 ganglioside. Ann Neurol. 2001;49:712–20.PubMedCrossRef
44.
go back to reference Susuki K, Yuki N, Schafer DP, Hirata K, Zhang G, Funakoshi K, Rasband MN. Dysfunction of nodes of Ranvier: a mechanism for anti-ganglioside antibody-mediated neuropathies. Exp Neurol. 2012;233:534–42.PubMedCrossRef Susuki K, Yuki N, Schafer DP, Hirata K, Zhang G, Funakoshi K, Rasband MN. Dysfunction of nodes of Ranvier: a mechanism for anti-ganglioside antibody-mediated neuropathies. Exp Neurol. 2012;233:534–42.PubMedCrossRef
45.
go back to reference Uncini A, Susuki K, Yuki N. Nodo-paranodopathy: beyond the demyelinating and axonal classification in anti-ganglioside antibody-mediated neuropathies. Clin Neurophysiol. 2013;124:1928–34.PubMedCrossRef Uncini A, Susuki K, Yuki N. Nodo-paranodopathy: beyond the demyelinating and axonal classification in anti-ganglioside antibody-mediated neuropathies. Clin Neurophysiol. 2013;124:1928–34.PubMedCrossRef
46.
go back to reference Bowes T, Wagner ER, Boffey J, Nicholl D, Cochrane L, Benboubetra M, Conner J, Furukawa K, Furukawa K, Willison HJ. Tolerance to self gangliosides is the major factor restricting the antibody response to lipopolysaccharide core oligosaccharides in campylobacter jejuni strains associated with Guillain-Barre syndrome. Infect Immun. 2002;70:5008–18.PubMedPubMedCentralCrossRef Bowes T, Wagner ER, Boffey J, Nicholl D, Cochrane L, Benboubetra M, Conner J, Furukawa K, Furukawa K, Willison HJ. Tolerance to self gangliosides is the major factor restricting the antibody response to lipopolysaccharide core oligosaccharides in campylobacter jejuni strains associated with Guillain-Barre syndrome. Infect Immun. 2002;70:5008–18.PubMedPubMedCentralCrossRef
47.
go back to reference Lunn MP, Johnson LA, Fromholt SE, Itonori S, Huang J, Vyas AA, Hildreth JE, Griffin JW, Schnaar RL, Sheikh KA. High-affinity anti-ganglioside IgG antibodies raised in complex ganglioside knockout mice: reexamination of GD1a immunolocalization. J Neurochem. 2000;75:404–12.PubMedCrossRef Lunn MP, Johnson LA, Fromholt SE, Itonori S, Huang J, Vyas AA, Hildreth JE, Griffin JW, Schnaar RL, Sheikh KA. High-affinity anti-ganglioside IgG antibodies raised in complex ganglioside knockout mice: reexamination of GD1a immunolocalization. J Neurochem. 2000;75:404–12.PubMedCrossRef
48.
go back to reference Halstead SK, Zitman FM, Humphreys PD, Greenshields K, Verschuuren JJ, Jacobs BC, Rother RP, Plomp JJ, Willison HJ. Eculizumab prevents anti-ganglioside antibody-mediated neuropathy in a murine model. Brain. 2008;131:1197–208.PubMedCrossRef Halstead SK, Zitman FM, Humphreys PD, Greenshields K, Verschuuren JJ, Jacobs BC, Rother RP, Plomp JJ, Willison HJ. Eculizumab prevents anti-ganglioside antibody-mediated neuropathy in a murine model. Brain. 2008;131:1197–208.PubMedCrossRef
49.
go back to reference Hadden RD, Gregson NA, Gold R, Willison HJ, Hughes RA. Guillain-Barre syndrome serum and anti-campylobacter antibody do not exacerbate experimental autoimmune neuritis. J Neuroimmunol. 2001;119:306–16.PubMedCrossRef Hadden RD, Gregson NA, Gold R, Willison HJ, Hughes RA. Guillain-Barre syndrome serum and anti-campylobacter antibody do not exacerbate experimental autoimmune neuritis. J Neuroimmunol. 2001;119:306–16.PubMedCrossRef
50.
go back to reference Gabriel CM, Gregson NA, Hughes RA. Anti-PMP22 antibodies in patients with inflammatory neuropathy. J Neuroimmunol. 2000;104:139–46.PubMedCrossRef Gabriel CM, Gregson NA, Hughes RA. Anti-PMP22 antibodies in patients with inflammatory neuropathy. J Neuroimmunol. 2000;104:139–46.PubMedCrossRef
51.
go back to reference Csurhes PA, Sullivan AA, Green K, Pender MP, McCombe PA. T cell reactivity to P0, P2, PMP-22, and myelin basic protein in patients with Guillain-Barre syndrome and chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Neurosurg Psychiatry. 2005;76:1431–9.PubMedPubMedCentralCrossRef Csurhes PA, Sullivan AA, Green K, Pender MP, McCombe PA. T cell reactivity to P0, P2, PMP-22, and myelin basic protein in patients with Guillain-Barre syndrome and chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Neurosurg Psychiatry. 2005;76:1431–9.PubMedPubMedCentralCrossRef
52.
go back to reference Makowska A, Pritchard J, Sanvito L, Gregson N, Peakman M, Hayday A, Hughes R. Immune responses to myelin proteins in Guillain-Barre syndrome. J Neurol Neurosurg Psychiatry. 2008;79:664–71.PubMedCrossRef Makowska A, Pritchard J, Sanvito L, Gregson N, Peakman M, Hayday A, Hughes R. Immune responses to myelin proteins in Guillain-Barre syndrome. J Neurol Neurosurg Psychiatry. 2008;79:664–71.PubMedCrossRef
53.
go back to reference Inglis HR, Csurhes PA, McCombe PA. Antibody responses to peptides of peripheral nerve myelin proteins P0 and P2 in patients with inflammatory demyelinating neuropathy. J Neurol Neurosurg Psychiatry. 2007;78:419–22.PubMedCrossRef Inglis HR, Csurhes PA, McCombe PA. Antibody responses to peptides of peripheral nerve myelin proteins P0 and P2 in patients with inflammatory demyelinating neuropathy. J Neurol Neurosurg Psychiatry. 2007;78:419–22.PubMedCrossRef
54.
go back to reference Samukawa M, Kuwahara M, Morikawa M, Ueno R, Hamada Y, Takada K, Hirano M, Mitsui Y, Sonoo M, Kusunoki S. Electrophysiological assessment of Guillain-Barre syndrome with both Gal-C and ganglioside antibodies; tendency for demyelinating type. J Neuroimmunol. 2016;301:61–4.PubMedCrossRef Samukawa M, Kuwahara M, Morikawa M, Ueno R, Hamada Y, Takada K, Hirano M, Mitsui Y, Sonoo M, Kusunoki S. Electrophysiological assessment of Guillain-Barre syndrome with both Gal-C and ganglioside antibodies; tendency for demyelinating type. J Neuroimmunol. 2016;301:61–4.PubMedCrossRef
55.
go back to reference Kharwar NK, Prasad KN, Singh K, Paliwal VK, Modi DR. Polymorphisms of IL-17 and ICAM-1 and their expression in Guillain-Barre syndrome. Int J Neurosci. 2017;127:680–87. Kharwar NK, Prasad KN, Singh K, Paliwal VK, Modi DR. Polymorphisms of IL-17 and ICAM-1 and their expression in Guillain-Barre syndrome. Int J Neurosci. 2017;127:680–87.
56.
go back to reference Meyer zu Horste G, Mausberg AK, Cordes S, El-Haddad H, Partke HJ, Leussink VI, Roden M, Martin S, Steinman L, Hartung HP, Kieseier BC. Thymic epithelium determines a spontaneous chronic neuritis in Icam1(tm1Jcgr)NOD mice. J Immunol. 2014;193:2678–90.PubMedCrossRef Meyer zu Horste G, Mausberg AK, Cordes S, El-Haddad H, Partke HJ, Leussink VI, Roden M, Martin S, Steinman L, Hartung HP, Kieseier BC. Thymic epithelium determines a spontaneous chronic neuritis in Icam1(tm1Jcgr)NOD mice. J Immunol. 2014;193:2678–90.PubMedCrossRef
57.
go back to reference Meyer zu Horste G, Cordes S, Pfaff J, Mathys C, Mausberg AK, Bendszus M, Pham M, Hartung HP, Kieseier BC. Predicting the response to intravenous Immunoglobulins in an animal model of chronic neuritis. PLoS One. 2016;11:e0164099.PubMedPubMedCentralCrossRef Meyer zu Horste G, Cordes S, Pfaff J, Mathys C, Mausberg AK, Bendszus M, Pham M, Hartung HP, Kieseier BC. Predicting the response to intravenous Immunoglobulins in an animal model of chronic neuritis. PLoS One. 2016;11:e0164099.PubMedPubMedCentralCrossRef
58.
go back to reference Zephir H, Stojkovic T, Latour P, Lacour A, de Seze J, Outteryck O, Maurage CA, Monpeurt C, Chatelet P, Ovelacq E, Vermersch P. Relapsing demyelinating disease affecting both the central and peripheral nervous systems. J Neurol Neurosurg Psychiatry. 2008;79:1032–9.PubMedCrossRef Zephir H, Stojkovic T, Latour P, Lacour A, de Seze J, Outteryck O, Maurage CA, Monpeurt C, Chatelet P, Ovelacq E, Vermersch P. Relapsing demyelinating disease affecting both the central and peripheral nervous systems. J Neurol Neurosurg Psychiatry. 2008;79:1032–9.PubMedCrossRef
59.
go back to reference Khalili-Shirazi A, Atkinson P, Gregson N, Hughes RA. Antibody responses to P0 and P2 myelin proteins in Guillain-Barre syndrome and chronic idiopathic demyelinating polyradiculoneuropathy. J Neuroimmunol. 1993;46:245–51.PubMedCrossRef Khalili-Shirazi A, Atkinson P, Gregson N, Hughes RA. Antibody responses to P0 and P2 myelin proteins in Guillain-Barre syndrome and chronic idiopathic demyelinating polyradiculoneuropathy. J Neuroimmunol. 1993;46:245–51.PubMedCrossRef
60.
go back to reference Sanvito L, Makowska A, Mahdi-Rogers M, Hadden RD, Peakman M, Gregson N, Nemni R, Hughes RA. Humoral and cellular immune responses to myelin protein peptides in chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Neurosurg Psychiatry. 2009;80:333–8.PubMedCrossRef Sanvito L, Makowska A, Mahdi-Rogers M, Hadden RD, Peakman M, Gregson N, Nemni R, Hughes RA. Humoral and cellular immune responses to myelin protein peptides in chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Neurosurg Psychiatry. 2009;80:333–8.PubMedCrossRef
61.
go back to reference Kwa MS, van Schaik IN, Brand A, Baas F, Vermeulen M. Investigation of serum response to PMP22, connexin 32 and P(0) in inflammatory neuropathies. J Neuroimmunol. 2001;116:220–5.PubMedCrossRef Kwa MS, van Schaik IN, Brand A, Baas F, Vermeulen M. Investigation of serum response to PMP22, connexin 32 and P(0) in inflammatory neuropathies. J Neuroimmunol. 2001;116:220–5.PubMedCrossRef
62.
go back to reference Kuwahara M, Suzuki S, Takada K, Kusunoki S. Antibodies to LM1 and LM1-containing ganglioside complexes in Guillain-Barre syndrome and chronic inflammatory demyelinating polyneuropathy. J Neuroimmunol. 2011;239:87–90.PubMedCrossRef Kuwahara M, Suzuki S, Takada K, Kusunoki S. Antibodies to LM1 and LM1-containing ganglioside complexes in Guillain-Barre syndrome and chronic inflammatory demyelinating polyneuropathy. J Neuroimmunol. 2011;239:87–90.PubMedCrossRef
63.
go back to reference Pollard JD, Armati PJ. CIDP - the relevance of recent advances in Schwann cell/axonal neurobiology. J Peripher Nerv Syst. 2011;16:15–23.PubMedCrossRef Pollard JD, Armati PJ. CIDP - the relevance of recent advances in Schwann cell/axonal neurobiology. J Peripher Nerv Syst. 2011;16:15–23.PubMedCrossRef
64.
go back to reference Querol L, Nogales-Gadea G, Rojas-Garcia R, Diaz-Manera J, Pardo J, Ortega-Moreno A, Sedano MJ, Gallardo E, Berciano J, Blesa R, et al. Neurofascin IgG4 antibodies in CIDP associate with disabling tremor and poor response to IVIg. Neurology. 2014;82:879–86.PubMedPubMedCentralCrossRef Querol L, Nogales-Gadea G, Rojas-Garcia R, Diaz-Manera J, Pardo J, Ortega-Moreno A, Sedano MJ, Gallardo E, Berciano J, Blesa R, et al. Neurofascin IgG4 antibodies in CIDP associate with disabling tremor and poor response to IVIg. Neurology. 2014;82:879–86.PubMedPubMedCentralCrossRef
65.
go back to reference Devaux JJ, Miura Y, Fukami Y, Inoue T, Manso C, Belghazi M, Sekiguchi K, Kokubun N, Ichikawa H, Wong AH, Yuki N. Neurofascin-155 IgG4 in chronic inflammatory demyelinating polyneuropathy. Neurology. 2016;86:800–7.PubMedPubMedCentralCrossRef Devaux JJ, Miura Y, Fukami Y, Inoue T, Manso C, Belghazi M, Sekiguchi K, Kokubun N, Ichikawa H, Wong AH, Yuki N. Neurofascin-155 IgG4 in chronic inflammatory demyelinating polyneuropathy. Neurology. 2016;86:800–7.PubMedPubMedCentralCrossRef
66.
go back to reference Delmont E, Manso C, Querol L, Cortese A, Berardinelli A, Lozza A, Belghazi M, Malissart P, Labauge P, Taieb G, et al. Autoantibodies to nodal isoforms of neurofascin in chronic inflammatory demyelinating polyneuropathy. Brain. 2017;140:1851–58. Delmont E, Manso C, Querol L, Cortese A, Berardinelli A, Lozza A, Belghazi M, Malissart P, Labauge P, Taieb G, et al. Autoantibodies to nodal isoforms of neurofascin in chronic inflammatory demyelinating polyneuropathy. Brain. 2017;140:1851–58.
67.
go back to reference Yan W, Nguyen T, Yuki N, Ji Q, Yiannikas C, Pollard JD, Mathey EK. Antibodies to neurofascin exacerbate adoptive transfer experimental autoimmune neuritis. J Neuroimmunol. 2014;277:13–7.PubMedCrossRef Yan W, Nguyen T, Yuki N, Ji Q, Yiannikas C, Pollard JD, Mathey EK. Antibodies to neurofascin exacerbate adoptive transfer experimental autoimmune neuritis. J Neuroimmunol. 2014;277:13–7.PubMedCrossRef
68.
go back to reference Mathey EK, Derfuss T, Storch MK, Williams KR, Hales K, Woolley DR, Al-Hayani A, Davies SN, Rasband MN, Olsson T, et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med. 2007;204:2363–72.PubMedPubMedCentralCrossRef Mathey EK, Derfuss T, Storch MK, Williams KR, Hales K, Woolley DR, Al-Hayani A, Davies SN, Rasband MN, Olsson T, et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med. 2007;204:2363–72.PubMedPubMedCentralCrossRef
69.
go back to reference Doppler K, Appeltshauser L, Wilhelmi K, Villmann C, Dib-Hajj SD, Waxman SG, Maurer M, Weishaupt A, Sommer C. Destruction of paranodal architecture in inflammatory neuropathy with anti-contactin-1 autoantibodies. J Neurol Neurosurg Psychiatry. 2015;86:720–8.PubMedCrossRef Doppler K, Appeltshauser L, Wilhelmi K, Villmann C, Dib-Hajj SD, Waxman SG, Maurer M, Weishaupt A, Sommer C. Destruction of paranodal architecture in inflammatory neuropathy with anti-contactin-1 autoantibodies. J Neurol Neurosurg Psychiatry. 2015;86:720–8.PubMedCrossRef
70.
go back to reference Manso C, Querol L, Mekaouche M, Illa I, Devaux JJ. Contactin-1 IgG4 antibodies cause paranode dismantling and conduction defects. Brain. 2016;139:1700–12.PubMedCrossRef Manso C, Querol L, Mekaouche M, Illa I, Devaux JJ. Contactin-1 IgG4 antibodies cause paranode dismantling and conduction defects. Brain. 2016;139:1700–12.PubMedCrossRef
71.
go back to reference Joshi AR, Holtmann L, Bobylev I, Schneider C, Ritter C, Weis J, Lehmann HC. Loss of Schwann cell plasticity in chronic inflammatory demyelinating polyneuropathy (CIDP). J Neuroinflammation. 2016;13:255.PubMedPubMedCentralCrossRef Joshi AR, Holtmann L, Bobylev I, Schneider C, Ritter C, Weis J, Lehmann HC. Loss of Schwann cell plasticity in chronic inflammatory demyelinating polyneuropathy (CIDP). J Neuroinflammation. 2016;13:255.PubMedPubMedCentralCrossRef
72.
go back to reference Lachance DH, Lennon VA, Pittock SJ, Tracy JA, Krecke KN, Amrami KK, Poeschla EM, Orenstein R, Scheithauer BW, Sejvar JJ, et al. An outbreak of neurological autoimmunity with polyradiculoneuropathy in workers exposed to aerosolised porcine neural tissue: a descriptive study. Lancet Neurol. 2010;9:55–66.PubMedCrossRef Lachance DH, Lennon VA, Pittock SJ, Tracy JA, Krecke KN, Amrami KK, Poeschla EM, Orenstein R, Scheithauer BW, Sejvar JJ, et al. An outbreak of neurological autoimmunity with polyradiculoneuropathy in workers exposed to aerosolised porcine neural tissue: a descriptive study. Lancet Neurol. 2010;9:55–66.PubMedCrossRef
73.
go back to reference St-Amour I, Pare I, Tremblay C, Coulombe K, Bazin R, Calon F. IVIg protects the 3xTg-AD mouse model of Alzheimer’s disease from memory deficit and Abeta pathology. J Neuroinflammation. 2014;11:54.PubMedPubMedCentralCrossRef St-Amour I, Pare I, Tremblay C, Coulombe K, Bazin R, Calon F. IVIg protects the 3xTg-AD mouse model of Alzheimer’s disease from memory deficit and Abeta pathology. J Neuroinflammation. 2014;11:54.PubMedPubMedCentralCrossRef
74.
go back to reference Schneider-Hohendorf T, Schwab N, Uceyler N, Gobel K, Sommer C, Wiendl H. CD8+ T-cell immunity in chronic inflammatory demyelinating polyradiculoneuropathy. Neurology. 2012;78:402–8.PubMedCrossRef Schneider-Hohendorf T, Schwab N, Uceyler N, Gobel K, Sommer C, Wiendl H. CD8+ T-cell immunity in chronic inflammatory demyelinating polyradiculoneuropathy. Neurology. 2012;78:402–8.PubMedCrossRef
75.
go back to reference Yang M, Rainone A, Shi XQ, Fournier S, Zhang J. A new animal model of spontaneous autoimmune peripheral polyneuropathy: implications for Guillain-Barre syndrome. Acta Neuropathol Commun. 2014;2:5.PubMedPubMedCentralCrossRef Yang M, Rainone A, Shi XQ, Fournier S, Zhang J. A new animal model of spontaneous autoimmune peripheral polyneuropathy: implications for Guillain-Barre syndrome. Acta Neuropathol Commun. 2014;2:5.PubMedPubMedCentralCrossRef
76.
go back to reference Waksman BH, Adams RD. Allergic neuritis: an experimental disease of rabbits induced by the injection of peripheral nervous tissue and adjuvants. J Exp Med. 1955;102:213–36.PubMedPubMedCentralCrossRef Waksman BH, Adams RD. Allergic neuritis: an experimental disease of rabbits induced by the injection of peripheral nervous tissue and adjuvants. J Exp Med. 1955;102:213–36.PubMedPubMedCentralCrossRef
77.
go back to reference Patzig J, Jahn O, Tenzer S, Wichert SP, de Monasterio-Schrader P, Rosfa S, Kuharev J, Yan K, Bormuth I, Bremer J, et al. Quantitative and integrative proteome analysis of peripheral nerve myelin identifies novel myelin proteins and candidate neuropathy loci. J Neurosci. 2011;31:16369–86.PubMedCrossRef Patzig J, Jahn O, Tenzer S, Wichert SP, de Monasterio-Schrader P, Rosfa S, Kuharev J, Yan K, Bormuth I, Bremer J, et al. Quantitative and integrative proteome analysis of peripheral nerve myelin identifies novel myelin proteins and candidate neuropathy loci. J Neurosci. 2011;31:16369–86.PubMedCrossRef
78.
go back to reference Gabriel CM, Hughes RA, Moore SE, Smith KJ, Walsh FS. Induction of experimental autoimmune neuritis with peripheral myelin protein-22. Brain. 1998;121(Pt 10):1895–902.PubMedCrossRef Gabriel CM, Hughes RA, Moore SE, Smith KJ, Walsh FS. Induction of experimental autoimmune neuritis with peripheral myelin protein-22. Brain. 1998;121(Pt 10):1895–902.PubMedCrossRef
79.
go back to reference Weerth S, Berger T, Lassmann H, Linington C. Encephalitogenic and neuritogenic T cell responses to the myelin-associated glycoprotein (MAG) in the Lewis rat. J Neuroimmunol. 1999;95:157–64.PubMedCrossRef Weerth S, Berger T, Lassmann H, Linington C. Encephalitogenic and neuritogenic T cell responses to the myelin-associated glycoprotein (MAG) in the Lewis rat. J Neuroimmunol. 1999;95:157–64.PubMedCrossRef
80.
go back to reference Gold R, Stoll G, Kieseier BC, Hartung HP, Toyka KV. Experimental Autoimmune Neuritis. In: Dyck PJ, Thomas PK, editors. Peripheral neuropathy, vol. 2. 4th ed. Philadelphia: Elsevier Saunders; 2005. Gold R, Stoll G, Kieseier BC, Hartung HP, Toyka KV. Experimental Autoimmune Neuritis. In: Dyck PJ, Thomas PK, editors. Peripheral neuropathy, vol. 2. 4th ed. Philadelphia: Elsevier Saunders; 2005.
81.
go back to reference Hartung HP, Schäfer B, Heininger K, Stoll G, Toyka KV. The role of macrophages and eicosanoids in the pathogenesis of experimental allergic neuritis. Serial clinical, electrophysiological, biochemical and morphological observations. Brain. 1988;111:1039–59.PubMedCrossRef Hartung HP, Schäfer B, Heininger K, Stoll G, Toyka KV. The role of macrophages and eicosanoids in the pathogenesis of experimental allergic neuritis. Serial clinical, electrophysiological, biochemical and morphological observations. Brain. 1988;111:1039–59.PubMedCrossRef
82.
go back to reference Prineas JW. Acute idiopathic polyneuritis. An electron microscope study. Lab Investig. 1972;26:133–47.PubMed Prineas JW. Acute idiopathic polyneuritis. An electron microscope study. Lab Investig. 1972;26:133–47.PubMed
83.
go back to reference St Charles JL, Bell JA, Gadsden BJ, Malik A, Cooke H, Van de Grift LK, Kim HY, Smith EJ, Mansfield LS: Guillain Barre syndrome is induced in non-obese diabetic (NOD) mice following campylobacter jejuni infection and is exacerbated by antibiotics. J Autoimmun. 2016;77:11–38. St Charles JL, Bell JA, Gadsden BJ, Malik A, Cooke H, Van de Grift LK, Kim HY, Smith EJ, Mansfield LS: Guillain Barre syndrome is induced in non-obese diabetic (NOD) mice following campylobacter jejuni infection and is exacerbated by antibiotics. J Autoimmun. 2016;77:11–38.
84.
go back to reference Wang Y, Shi Q, Lv H, Hu M, Wang W, Wang Q, Qiao B, Zhang G, Lv Z, Kjellman C, et al. IgG-degrading enzyme of Streptococcus pyogenes (IdeS) prevents disease progression and facilitates improvement in a rabbit model of Guillain-Barre syndrome. Exp Neurol. 2017;291:134–40.PubMedCrossRef Wang Y, Shi Q, Lv H, Hu M, Wang W, Wang Q, Qiao B, Zhang G, Lv Z, Kjellman C, et al. IgG-degrading enzyme of Streptococcus pyogenes (IdeS) prevents disease progression and facilitates improvement in a rabbit model of Guillain-Barre syndrome. Exp Neurol. 2017;291:134–40.PubMedCrossRef
85.
go back to reference Jung S, Gaupp S, Korn T, Kollner G, Hartung HP, Toyka KV. Biphasic form of experimental autoimmune neuritis in dark agouti rats and its oral therapy by antigen-specific tolerization. J Neurosci Res. 2004;75:524–35.PubMedCrossRef Jung S, Gaupp S, Korn T, Kollner G, Hartung HP, Toyka KV. Biphasic form of experimental autoimmune neuritis in dark agouti rats and its oral therapy by antigen-specific tolerization. J Neurosci Res. 2004;75:524–35.PubMedCrossRef
86.
go back to reference McCombe PA, Harness J, Pender MP. Effects of cyclosporin A treatment on clinical course and inflammatory cell apoptosis in experimental autoimmune encephalomyelitis induced in Lewis rats by inoculation with myelin basic protein. J Neuroimmunol. 1999;97:60–9.PubMedCrossRef McCombe PA, Harness J, Pender MP. Effects of cyclosporin A treatment on clinical course and inflammatory cell apoptosis in experimental autoimmune encephalomyelitis induced in Lewis rats by inoculation with myelin basic protein. J Neuroimmunol. 1999;97:60–9.PubMedCrossRef
87.
go back to reference McCombe PA, van der Kreek SA, Pender MP. Neuropathological findings in chronic relapsing experimental allergic neuritis induced in the Lewis rat by inoculation with intradural root myelin and treatment with low dose cyclosporin A. Neuropathol Appl Neurobiol. 1992;18:171–87.PubMedCrossRef McCombe PA, van der Kreek SA, Pender MP. Neuropathological findings in chronic relapsing experimental allergic neuritis induced in the Lewis rat by inoculation with intradural root myelin and treatment with low dose cyclosporin A. Neuropathol Appl Neurobiol. 1992;18:171–87.PubMedCrossRef
88.
go back to reference Harness J, Pender MP, McCombe PA. Cyclosporin a treatment modulates cytokine mRNA expression by inflammatory cells extracted from the spinal cord of rats with experimental autoimmune encephalomyelitis induced by inoculation with myelin basic protein. J Neurol Sci. 2001;187:7–16.PubMedCrossRef Harness J, Pender MP, McCombe PA. Cyclosporin a treatment modulates cytokine mRNA expression by inflammatory cells extracted from the spinal cord of rats with experimental autoimmune encephalomyelitis induced by inoculation with myelin basic protein. J Neurol Sci. 2001;187:7–16.PubMedCrossRef
89.
go back to reference Granelli-Piperno A, Keane M, Steinman RM. Evidence that cyclosporine inhibits cell-mediated immunity primarily at the level of the T lymphocyte rather than the accessory cell. Transplantation. 1988;46:53S–60S.PubMedCrossRef Granelli-Piperno A, Keane M, Steinman RM. Evidence that cyclosporine inhibits cell-mediated immunity primarily at the level of the T lymphocyte rather than the accessory cell. Transplantation. 1988;46:53S–60S.PubMedCrossRef
90.
go back to reference Saitoh K, Kon S, Nakatsuru T, Inui K, Ihara T, Matsumoto N, Kitai Y, Muromoto R, Matsuda T. Anti-IL-17A blocking antibody reduces cyclosporin A-induced relapse in experimental autoimmune encephalomyelitis mice. Biochem Biophys Rep. 2016;8:139–45.PubMedPubMedCentral Saitoh K, Kon S, Nakatsuru T, Inui K, Ihara T, Matsumoto N, Kitai Y, Muromoto R, Matsuda T. Anti-IL-17A blocking antibody reduces cyclosporin A-induced relapse in experimental autoimmune encephalomyelitis mice. Biochem Biophys Rep. 2016;8:139–45.PubMedPubMedCentral
91.
go back to reference Kang HG, Zhang D, Degauque N, Mariat C, Alexopoulos S, Zheng XX. Effects of cyclosporine on transplant tolerance: the role of IL-2. Am J Transplant. 2007;7:1907–16.PubMedCrossRef Kang HG, Zhang D, Degauque N, Mariat C, Alexopoulos S, Zheng XX. Effects of cyclosporine on transplant tolerance: the role of IL-2. Am J Transplant. 2007;7:1907–16.PubMedCrossRef
92.
go back to reference Almeida AR, Legrand N, Papiernik M, Freitas AA. Homeostasis of peripheral CD4+ T cells: IL-2R alpha and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers. J Immunol. 2002;169:4850–60.PubMedCrossRef Almeida AR, Legrand N, Papiernik M, Freitas AA. Homeostasis of peripheral CD4+ T cells: IL-2R alpha and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers. J Immunol. 2002;169:4850–60.PubMedCrossRef
93.
go back to reference Miroux C, Morales O, Ghazal K, Othman SB, de Launoit Y, Pancre V, Conti F, Delhem N. In vitro effects of cyclosporine A and tacrolimus on regulatory T-cell proliferation and function. Transplantation. 2012;94:123–31.PubMedCrossRef Miroux C, Morales O, Ghazal K, Othman SB, de Launoit Y, Pancre V, Conti F, Delhem N. In vitro effects of cyclosporine A and tacrolimus on regulatory T-cell proliferation and function. Transplantation. 2012;94:123–31.PubMedCrossRef
94.
go back to reference Lassmann H, Fierz W, Neuchrist C, Meyermann R. Chronic relapsing experimental allergic neuritis induced by repeated transfer of P2-protein reactive T cell lines. Brain. 1991;114(Pt 1B):429–42.PubMedCrossRef Lassmann H, Fierz W, Neuchrist C, Meyermann R. Chronic relapsing experimental allergic neuritis induced by repeated transfer of P2-protein reactive T cell lines. Brain. 1991;114(Pt 1B):429–42.PubMedCrossRef
95.
go back to reference Zehntner SP, Brisebois M, Tran E, Owens T, Fournier S. Constitutive expression of a costimulatory ligand on antigen-presenting cells in the nervous system drives demyelinating disease. FASEB J. 2003;17:1910–2.PubMed Zehntner SP, Brisebois M, Tran E, Owens T, Fournier S. Constitutive expression of a costimulatory ligand on antigen-presenting cells in the nervous system drives demyelinating disease. FASEB J. 2003;17:1910–2.PubMed
96.
go back to reference Brisebois M, Zehntner SP, Estrada J, Owens T, Fournier S. A pathogenic role for CD8+ T cells in a spontaneous model of demyelinating disease. J Immunol. 2006;177:2403–11.PubMedCrossRef Brisebois M, Zehntner SP, Estrada J, Owens T, Fournier S. A pathogenic role for CD8+ T cells in a spontaneous model of demyelinating disease. J Immunol. 2006;177:2403–11.PubMedCrossRef
97.
go back to reference Dace DS, Khan AA, Stark JL, Kelly J, Cross AH, Apte RS. Interleukin-10 overexpression promotes Fas-ligand-dependent chronic macrophage-mediated demyelinating polyneuropathy. PLoS One. 2009;4:e7121.PubMedPubMedCentralCrossRef Dace DS, Khan AA, Stark JL, Kelly J, Cross AH, Apte RS. Interleukin-10 overexpression promotes Fas-ligand-dependent chronic macrophage-mediated demyelinating polyneuropathy. PLoS One. 2009;4:e7121.PubMedPubMedCentralCrossRef
98.
go back to reference Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol. 2005;23:447–85.PubMedCrossRef Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol. 2005;23:447–85.PubMedCrossRef
99.
go back to reference Salomon B, Rhee L, Bour-Jordan H, Hsin H, Montag A, Soliven B, Arcella J, Girvin AM, Padilla J, Miller SD, Bluestone JA. Development of spontaneous autoimmune peripheral polyneuropathy in B7-2-deficient NOD mice. J Exp Med. 2001;194:677–84.PubMedPubMedCentralCrossRef Salomon B, Rhee L, Bour-Jordan H, Hsin H, Montag A, Soliven B, Arcella J, Girvin AM, Padilla J, Miller SD, Bluestone JA. Development of spontaneous autoimmune peripheral polyneuropathy in B7-2-deficient NOD mice. J Exp Med. 2001;194:677–84.PubMedPubMedCentralCrossRef
100.
go back to reference Bour-Jordan H, Thompson HL, Bluestone JA. Distinct effector mechanisms in the development of autoimmune neuropathy versus diabetes in nonobese diabetic mice. J Immunol. 2005;175:5649–55.PubMedCrossRef Bour-Jordan H, Thompson HL, Bluestone JA. Distinct effector mechanisms in the development of autoimmune neuropathy versus diabetes in nonobese diabetic mice. J Immunol. 2005;175:5649–55.PubMedCrossRef
101.
go back to reference Kim HJ, Jung CG, Jensen MA, Dukala D, Soliven B. Targeting of myelin protein zero in a spontaneous autoimmune polyneuropathy. J Immunol. 2008;181:8753–60.PubMedPubMedCentralCrossRef Kim HJ, Jung CG, Jensen MA, Dukala D, Soliven B. Targeting of myelin protein zero in a spontaneous autoimmune polyneuropathy. J Immunol. 2008;181:8753–60.PubMedPubMedCentralCrossRef
102.
go back to reference Louvet C, Kabre BG, Davini DW, Martinier N, Su MA, DeVoss JJ, Rosenthal WL, Anderson MS, Bour-Jordan H, Bluestone JA. A novel myelin P0-specific T cell receptor transgenic mouse develops a fulminant autoimmune peripheral neuropathy. J Exp Med. 2009;206:507–14.PubMedPubMedCentralCrossRef Louvet C, Kabre BG, Davini DW, Martinier N, Su MA, DeVoss JJ, Rosenthal WL, Anderson MS, Bour-Jordan H, Bluestone JA. A novel myelin P0-specific T cell receptor transgenic mouse develops a fulminant autoimmune peripheral neuropathy. J Exp Med. 2009;206:507–14.PubMedPubMedCentralCrossRef
103.
go back to reference Abraham PM, Quan SH, Dukala D, Soliven B. CD19 as a therapeutic target in a spontaneous autoimmune polyneuropathy. Clin Exp Immunol. 2014;175:181–91.PubMedPubMedCentralCrossRef Abraham PM, Quan SH, Dukala D, Soliven B. CD19 as a therapeutic target in a spontaneous autoimmune polyneuropathy. Clin Exp Immunol. 2014;175:181–91.PubMedPubMedCentralCrossRef
104.
go back to reference Kim HJ, Jung CG, Dukala D, Bae H, Kakazu R, Wollmann R, Soliven B. Fingolimod and related compounds in a spontaneous autoimmune polyneuropathy. J Neuroimmunol. 2009;214:93–100.PubMedPubMedCentralCrossRef Kim HJ, Jung CG, Dukala D, Bae H, Kakazu R, Wollmann R, Soliven B. Fingolimod and related compounds in a spontaneous autoimmune polyneuropathy. J Neuroimmunol. 2009;214:93–100.PubMedPubMedCentralCrossRef
105.
go back to reference Quan S, Sheng JR, Abraham PM, Soliven B. Regulatory T and B lymphocytes in a spontaneous autoimmune polyneuropathy. Clin Exp Immunol. 2016;184:50–61.PubMedPubMedCentralCrossRef Quan S, Sheng JR, Abraham PM, Soliven B. Regulatory T and B lymphocytes in a spontaneous autoimmune polyneuropathy. Clin Exp Immunol. 2016;184:50–61.PubMedPubMedCentralCrossRef
106.
go back to reference Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005;201:723–35.PubMedPubMedCentralCrossRef Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005;201:723–35.PubMedPubMedCentralCrossRef
107.
go back to reference Meyer zu Horste G, Cordes S, Mausberg AK, Zozulya AL, Wessig C, Sparwasser T, Mathys C, Wiendl H, Hartung HP, Kieseier BC. FoxP3+ regulatory T cells determine disease severity in rodent models of inflammatory neuropathies. PLoS One. 2014;e108756:9. Meyer zu Horste G, Cordes S, Mausberg AK, Zozulya AL, Wessig C, Sparwasser T, Mathys C, Wiendl H, Hartung HP, Kieseier BC. FoxP3+ regulatory T cells determine disease severity in rodent models of inflammatory neuropathies. PLoS One. 2014;e108756:9.
108.
go back to reference Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1−/− mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A. 2005;102:11823–8.PubMedPubMedCentralCrossRef Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1−/− mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A. 2005;102:11823–8.PubMedPubMedCentralCrossRef
109.
go back to reference Jiang F, Yoshida T, Nakaki F, Terawaki S, Chikuma S, Kato Y, Okazaki IM, Honjo T, Okazaki T. Identification of QTLs that modify peripheral neuropathy in NOD.H2b-Pdcd1−/− mice. Int Immunol. 2009;21:499–509.PubMedCrossRef Jiang F, Yoshida T, Nakaki F, Terawaki S, Chikuma S, Kato Y, Okazaki IM, Honjo T, Okazaki T. Identification of QTLs that modify peripheral neuropathy in NOD.H2b-Pdcd1−/− mice. Int Immunol. 2009;21:499–509.PubMedCrossRef
110.
go back to reference Yoshida T, Jiang F, Honjo T, Okazaki T. PD-1 deficiency reveals various tissue-specific autoimmunity by H-2b and dose-dependent requirement of H-2g7 for diabetes in NOD mice. Proc Natl Acad Sci U S A. 2008;105:3533–8.PubMedPubMedCentralCrossRef Yoshida T, Jiang F, Honjo T, Okazaki T. PD-1 deficiency reveals various tissue-specific autoimmunity by H-2b and dose-dependent requirement of H-2g7 for diabetes in NOD mice. Proc Natl Acad Sci U S A. 2008;105:3533–8.PubMedPubMedCentralCrossRef
111.
go back to reference Prevot N, Briet C, Lassmann H, Tardivel I, Roy E, Morin J, Mak TW, Tafuri A, Boitard C. Abrogation of ICOS/ICOS ligand costimulation in NOD mice results in autoimmune deviation toward the neuromuscular system. Eur J Immunol. 2010;40:2267–76.PubMedCrossRef Prevot N, Briet C, Lassmann H, Tardivel I, Roy E, Morin J, Mak TW, Tafuri A, Boitard C. Abrogation of ICOS/ICOS ligand costimulation in NOD mice results in autoimmune deviation toward the neuromuscular system. Eur J Immunol. 2010;40:2267–76.PubMedCrossRef
112.
go back to reference Zeng XL, Nagavalli A, Smith CJ, Howard JF, Su MA. Divergent effects of T cell costimulation and inflammatory cytokine production on autoimmune peripheral neuropathy provoked by Aire deficiency. J Immunol. 2013;190:3895–904.PubMedPubMedCentralCrossRef Zeng XL, Nagavalli A, Smith CJ, Howard JF, Su MA. Divergent effects of T cell costimulation and inflammatory cytokine production on autoimmune peripheral neuropathy provoked by Aire deficiency. J Immunol. 2013;190:3895–904.PubMedPubMedCentralCrossRef
113.
go back to reference Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517.PubMedCrossRef Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517.PubMedCrossRef
114.
go back to reference Vinuesa CG, Linterman MA, Yu D, MacLennan IC. Follicular helper T cells. Annu Rev Immunol. 2016;34:335–68.PubMedCrossRef Vinuesa CG, Linterman MA, Yu D, MacLennan IC. Follicular helper T cells. Annu Rev Immunol. 2016;34:335–68.PubMedCrossRef
115.
go back to reference Wang X, Zheng XY, Ma C, Wang XK, Wu J, Adem A, Zhu J, Zhang HL. Mitigated Tregs and augmented Th17 cells and cytokines are associated with severity of experimental autoimmune neuritis. Scand J Immunol. 2014;80:180–90.PubMedCrossRef Wang X, Zheng XY, Ma C, Wang XK, Wu J, Adem A, Zhu J, Zhang HL. Mitigated Tregs and augmented Th17 cells and cytokines are associated with severity of experimental autoimmune neuritis. Scand J Immunol. 2014;80:180–90.PubMedCrossRef
116.
go back to reference Li XL, Dou YC, Liu Y, Shi CW, Cao LL, Zhang XQ, Zhu J, Duan RS. Atorvastatin ameliorates experimental autoimmune neuritis by decreased Th1/Th17 cytokines and up-regulated T regulatory cells. Cell Immunol. 2011;271:455–61.PubMedCrossRef Li XL, Dou YC, Liu Y, Shi CW, Cao LL, Zhang XQ, Zhu J, Duan RS. Atorvastatin ameliorates experimental autoimmune neuritis by decreased Th1/Th17 cytokines and up-regulated T regulatory cells. Cell Immunol. 2011;271:455–61.PubMedCrossRef
117.
go back to reference Zhang Z, Zhang ZY, Fauser U, Schluesener HJ. FTY720 ameliorates experimental autoimmune neuritis by inhibition of lymphocyte and monocyte infiltration into peripheral nerves. Exp Neurol. 2008;210:681–90.PubMedCrossRef Zhang Z, Zhang ZY, Fauser U, Schluesener HJ. FTY720 ameliorates experimental autoimmune neuritis by inhibition of lymphocyte and monocyte infiltration into peripheral nerves. Exp Neurol. 2008;210:681–90.PubMedCrossRef
118.
go back to reference Chi LJ, Xu WH, Zhang ZW, Huang HT, Zhang LM, Zhou J. Distribution of Th17 cells and Th1 cells in peripheral blood and cerebrospinal fluid in chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2010;15:345–56.PubMedCrossRef Chi LJ, Xu WH, Zhang ZW, Huang HT, Zhang LM, Zhou J. Distribution of Th17 cells and Th1 cells in peripheral blood and cerebrospinal fluid in chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2010;15:345–56.PubMedCrossRef
119.
go back to reference Mei FJ, Ishizu T, Murai H, Osoegawa M, Minohara M, Zhang KN, Kira J. Th1 shift in CIDP versus Th2 shift in vasculitic neuropathy in CSF. J Neurol Sci. 2005;228:75–85.PubMedCrossRef Mei FJ, Ishizu T, Murai H, Osoegawa M, Minohara M, Zhang KN, Kira J. Th1 shift in CIDP versus Th2 shift in vasculitic neuropathy in CSF. J Neurol Sci. 2005;228:75–85.PubMedCrossRef
120.
go back to reference Madia F, Frisullo G, Nociti V, Conte A, Luigetti M, Del Grande A, Patanella AK, Iorio R, Tonali PA, Batocchi AP, Sabatelli M. pSTAT1, pSTAT3, and T-bet as markers of disease activity in chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2009;14:107–17.PubMedCrossRef Madia F, Frisullo G, Nociti V, Conte A, Luigetti M, Del Grande A, Patanella AK, Iorio R, Tonali PA, Batocchi AP, Sabatelli M. pSTAT1, pSTAT3, and T-bet as markers of disease activity in chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2009;14:107–17.PubMedCrossRef
121.
go back to reference Li S, Yu M, Li H, Zhang H, Jiang Y. IL-17 and IL-22 in cerebrospinal fluid and plasma are elevated in Guillain-Barre syndrome. Mediat Inflamm. 2012;2012:260473. Li S, Yu M, Li H, Zhang H, Jiang Y. IL-17 and IL-22 in cerebrospinal fluid and plasma are elevated in Guillain-Barre syndrome. Mediat Inflamm. 2012;2012:260473.
122.
go back to reference Li S, Jin T, Zhang HL, Yu H, Meng F, Concha Quezada H, Zhu J. Circulating Th17, Th22, and Th1 cells are elevated in the Guillain-Barre syndrome and downregulated by IVIg treatments. Mediat Inflamm. 2014;2014:740947. Li S, Jin T, Zhang HL, Yu H, Meng F, Concha Quezada H, Zhu J. Circulating Th17, Th22, and Th1 cells are elevated in the Guillain-Barre syndrome and downregulated by IVIg treatments. Mediat Inflamm. 2014;2014:740947.
123.
go back to reference Huh JR, Leung MW, Huang P, Ryan DA, Krout MR, Malapaka RR, Chow J, Manel N, Ciofani M, Kim SV, et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORgammat activity. Nature. 2011;472:486–90.PubMedPubMedCentralCrossRef Huh JR, Leung MW, Huang P, Ryan DA, Krout MR, Malapaka RR, Chow J, Manel N, Ciofani M, Kim SV, et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORgammat activity. Nature. 2011;472:486–90.PubMedPubMedCentralCrossRef
124.
go back to reference Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, Liu J, Istrate MA, Kamenecka TM, Roush WR, Vidovic D, et al. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature. 2011;472:491–4.PubMedPubMedCentralCrossRef Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, Liu J, Istrate MA, Kamenecka TM, Roush WR, Vidovic D, et al. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature. 2011;472:491–4.PubMedPubMedCentralCrossRef
126.
go back to reference Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, Levy-Lahad E, Mazzella M, Goulet O, Perroni L, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20.PubMedCrossRef Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, Levy-Lahad E, Mazzella M, Goulet O, Perroni L, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20.PubMedCrossRef
127.
go back to reference Buckner JH. Mechanisms of impaired regulation by CD4(+)CD25(+)FOXP3(+) regulatory T cells in human autoimmune diseases. Nat Rev Immunol. 2010;10:849–59.PubMedPubMedCentralCrossRef Buckner JH. Mechanisms of impaired regulation by CD4(+)CD25(+)FOXP3(+) regulatory T cells in human autoimmune diseases. Nat Rev Immunol. 2010;10:849–59.PubMedPubMedCentralCrossRef
128.
go back to reference Chi LJ, Wang HB, Wang WZ. Impairment of circulating CD4+CD25+ regulatory T cells in patients with chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2008;13:54–63.PubMedCrossRef Chi LJ, Wang HB, Wang WZ. Impairment of circulating CD4+CD25+ regulatory T cells in patients with chronic inflammatory demyelinating polyradiculoneuropathy. J Peripher Nerv Syst. 2008;13:54–63.PubMedCrossRef
129.
go back to reference Harness J, McCombe PA. Increased levels of activated T-cells and reduced levels of CD4/CD25+ cells in peripheral blood of Guillain-Barre syndrome patients compared to controls. J Clin Neurosci. 2008;15:1031–5.PubMedCrossRef Harness J, McCombe PA. Increased levels of activated T-cells and reduced levels of CD4/CD25+ cells in peripheral blood of Guillain-Barre syndrome patients compared to controls. J Clin Neurosci. 2008;15:1031–5.PubMedCrossRef
130.
go back to reference Zhang Z, Zhang ZY, Fauser U, Schluesener HJ. Distribution of Foxp3(+) T-regulatory cells in experimental autoimmune neuritis rats. Exp Neurol. 2009;216:75–82.PubMedCrossRef Zhang Z, Zhang ZY, Fauser U, Schluesener HJ. Distribution of Foxp3(+) T-regulatory cells in experimental autoimmune neuritis rats. Exp Neurol. 2009;216:75–82.PubMedCrossRef
131.
go back to reference Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6.PubMedCrossRef Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6.PubMedCrossRef
132.
go back to reference Maddur MS, Trinath J, Rabin M, Bolgert F, Guy M, Vallat JM, Magy L, Balaji KN, Kaveri SV, Bayry J. Intravenous immunoglobulin-mediated expansion of regulatory T cells in autoimmune patients is associated with increased prostaglandin E2 levels in the circulation. Cell Mol Immunol. 2015;12:650–2.PubMedCrossRef Maddur MS, Trinath J, Rabin M, Bolgert F, Guy M, Vallat JM, Magy L, Balaji KN, Kaveri SV, Bayry J. Intravenous immunoglobulin-mediated expansion of regulatory T cells in autoimmune patients is associated with increased prostaglandin E2 levels in the circulation. Cell Mol Immunol. 2015;12:650–2.PubMedCrossRef
133.
go back to reference Fantini MC, Dominitzki S, Rizzo A, Neurath MF, Becker C. In vitro generation of CD4+ CD25+ regulatory cells from murine naive T cells. Nat Protoc. 2007;2:1789–94.PubMedCrossRef Fantini MC, Dominitzki S, Rizzo A, Neurath MF, Becker C. In vitro generation of CD4+ CD25+ regulatory cells from murine naive T cells. Nat Protoc. 2007;2:1789–94.PubMedCrossRef
134.
go back to reference Calik MW, Shankarappa SA, Langert KA, Stubbs EB Jr. Forced exercise preconditioning attenuates experimental autoimmune neuritis by altering Th1 lymphocyte composition and egress. ASN Neuro. 2015;7. Calik MW, Shankarappa SA, Langert KA, Stubbs EB Jr. Forced exercise preconditioning attenuates experimental autoimmune neuritis by altering Th1 lymphocyte composition and egress. ASN Neuro. 2015;7.
135.
go back to reference Ding Y, Han R, Jiang W, Xiao J, Liu H, Chen X, Li X, Hao J. Programmed death Ligand 1 plays a Neuroprotective role in experimental autoimmune neuritis by controlling peripheral nervous system inflammation of rats. J Immunol. 2016;197:3831–40.PubMedCrossRef Ding Y, Han R, Jiang W, Xiao J, Liu H, Chen X, Li X, Hao J. Programmed death Ligand 1 plays a Neuroprotective role in experimental autoimmune neuritis by controlling peripheral nervous system inflammation of rats. J Immunol. 2016;197:3831–40.PubMedCrossRef
136.
go back to reference Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–89.PubMedCrossRef Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–89.PubMedCrossRef
137.
go back to reference Yosef N, Ubogu EE. alpha(M)beta(2)-integrin-intercellular adhesion molecule-1 interactions drive the flow-dependent trafficking of Guillain-Barre syndrome patient derived mononuclear leukocytes at the blood-nerve barrier in vitro. J Cell Physiol. 2012;227:3857–75.PubMedPubMedCentralCrossRef Yosef N, Ubogu EE. alpha(M)beta(2)-integrin-intercellular adhesion molecule-1 interactions drive the flow-dependent trafficking of Guillain-Barre syndrome patient derived mononuclear leukocytes at the blood-nerve barrier in vitro. J Cell Physiol. 2012;227:3857–75.PubMedPubMedCentralCrossRef
138.
go back to reference Van Steenwinckel J, Auvynet C, Sapienza A, Reaux-Le Goazigo A, Combadiere C, Melik Parsadaniantz S. Stromal cell-derived CCL2 drives neuropathic pain states through myeloid cell infiltration in injured nerve. Brain Behav Immun. 2015;45:198–210.PubMedCrossRef Van Steenwinckel J, Auvynet C, Sapienza A, Reaux-Le Goazigo A, Combadiere C, Melik Parsadaniantz S. Stromal cell-derived CCL2 drives neuropathic pain states through myeloid cell infiltration in injured nerve. Brain Behav Immun. 2015;45:198–210.PubMedCrossRef
139.
go back to reference Dong C, Palladino SP, Helton ES, Ubogu EE. The pathogenic relevance of alphaM-integrin in Guillain-Barre syndrome. Acta Neuropathol. 2016;132:739–52.PubMedPubMedCentralCrossRef Dong C, Palladino SP, Helton ES, Ubogu EE. The pathogenic relevance of alphaM-integrin in Guillain-Barre syndrome. Acta Neuropathol. 2016;132:739–52.PubMedPubMedCentralCrossRef
140.
go back to reference Archelos JJ, Maurer M, Jung S, Miyasaka M, Tamatani T, Toyka KV, Hartung HP. Inhibition of experimental autoimmune neuritis by an antibody to the lymphocyte function-associated antigen-1. Lab Investig. 1994;70:667–75.PubMed Archelos JJ, Maurer M, Jung S, Miyasaka M, Tamatani T, Toyka KV, Hartung HP. Inhibition of experimental autoimmune neuritis by an antibody to the lymphocyte function-associated antigen-1. Lab Investig. 1994;70:667–75.PubMed
141.
go back to reference Archelos JJ, Maurer M, Jung S, Toyka KV, Hartung HP. Suppression of experimental allergic neuritis by an antibody to the intracellular adhesion molecule ICAM-1. Brain. 1993;116(Pt 5):1043–58.PubMedCrossRef Archelos JJ, Maurer M, Jung S, Toyka KV, Hartung HP. Suppression of experimental allergic neuritis by an antibody to the intracellular adhesion molecule ICAM-1. Brain. 1993;116(Pt 5):1043–58.PubMedCrossRef
142.
go back to reference Xia RH, Yosef N, Ubogu EE. Selective expression and cellular localization of pro-inflammatory chemokine ligand/receptor pairs in the sciatic nerves of a severe murine experimental autoimmune neuritis model of Guillain-Barre syndrome. Neuropathol Appl Neurobiol. 2010;36:388–98.PubMedCrossRef Xia RH, Yosef N, Ubogu EE. Selective expression and cellular localization of pro-inflammatory chemokine ligand/receptor pairs in the sciatic nerves of a severe murine experimental autoimmune neuritis model of Guillain-Barre syndrome. Neuropathol Appl Neurobiol. 2010;36:388–98.PubMedCrossRef
143.
go back to reference Kieseier BC, Tani M, Mahad D, Oka N, Ho T, Woodroofe N, Griffin JW, Toyka KV, Ransohoff RM, Hartung HP. Chemokines and chemokine receptors in inflammatory demyelinating neuropathies: a central role for IP-10. Brain. 2002;125:823–34.PubMedCrossRef Kieseier BC, Tani M, Mahad D, Oka N, Ho T, Woodroofe N, Griffin JW, Toyka KV, Ransohoff RM, Hartung HP. Chemokines and chemokine receptors in inflammatory demyelinating neuropathies: a central role for IP-10. Brain. 2002;125:823–34.PubMedCrossRef
144.
go back to reference Orlikowski D, Chazaud B, Plonquet A, Poron F, Sharshar T, Maison P, Raphael JC, Gherardi RK, Creange A. Monocyte chemoattractant protein 1 and chemokine receptor CCR2 productions in Guillain-Barre syndrome and experimental autoimmune neuritis. J Neuroimmunol. 2003;134:118–27.PubMedCrossRef Orlikowski D, Chazaud B, Plonquet A, Poron F, Sharshar T, Maison P, Raphael JC, Gherardi RK, Creange A. Monocyte chemoattractant protein 1 and chemokine receptor CCR2 productions in Guillain-Barre syndrome and experimental autoimmune neuritis. J Neuroimmunol. 2003;134:118–27.PubMedCrossRef
145.
go back to reference Duan RS, Chen Z, Bao L, Quezada HC, Nennesmo I, Winblad B, Zhu J. CCR5 deficiency does not prevent P0 peptide 180-199 immunized mice from experimental autoimmune neuritis. Neurobiol Dis. 2004;16:630–7.PubMedCrossRef Duan RS, Chen Z, Bao L, Quezada HC, Nennesmo I, Winblad B, Zhu J. CCR5 deficiency does not prevent P0 peptide 180-199 immunized mice from experimental autoimmune neuritis. Neurobiol Dis. 2004;16:630–7.PubMedCrossRef
146.
go back to reference Brunn A, Utermohlen O, Mihelcic M, Sanchez-Ruiz M, Carstov M, Blau T, Ustinova I, Penfold M, Montesinos-Rongen M, Deckert M. Differential effects of CXCR4-CXCL12- and CXCR7-CXCL12-mediated immune reactions on murine P0106-125 -induced experimental autoimmune neuritis. Neuropathol Appl Neurobiol. 2013;39:772–87.PubMedCrossRef Brunn A, Utermohlen O, Mihelcic M, Sanchez-Ruiz M, Carstov M, Blau T, Ustinova I, Penfold M, Montesinos-Rongen M, Deckert M. Differential effects of CXCR4-CXCL12- and CXCR7-CXCL12-mediated immune reactions on murine P0106-125 -induced experimental autoimmune neuritis. Neuropathol Appl Neurobiol. 2013;39:772–87.PubMedCrossRef
147.
go back to reference Dong C, Greathouse KM, Beacham RL, Palladino SP, Helton ES, Ubogu EE. Fibronectin connecting segment-1 peptide inhibits pathogenic leukocyte trafficking and inflammatory demyelination in experimental models of chronic inflammatory demyelinating polyradiculoneuropathy. Exp Neurol. 2017;292:35–45.PubMedCrossRef Dong C, Greathouse KM, Beacham RL, Palladino SP, Helton ES, Ubogu EE. Fibronectin connecting segment-1 peptide inhibits pathogenic leukocyte trafficking and inflammatory demyelination in experimental models of chronic inflammatory demyelinating polyradiculoneuropathy. Exp Neurol. 2017;292:35–45.PubMedCrossRef
148.
go back to reference Greenwood J, Walters CE, Pryce G, Kanuga N, Beraud E, Baker D, Adamson P. Lovastatin inhibits brain endothelial cell rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. FASEB J. 2003;17:905–7.PubMed Greenwood J, Walters CE, Pryce G, Kanuga N, Beraud E, Baker D, Adamson P. Lovastatin inhibits brain endothelial cell rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. FASEB J. 2003;17:905–7.PubMed
149.
go back to reference Danesh FR, Anel RL, Zeng L, Lomasney J, Sahai A, Kanwar YS. Immunomodulatory effects of HMG-CoA reductase inhibitors. Arch Immunol Ther Exp. 2003;51:139–48. Danesh FR, Anel RL, Zeng L, Lomasney J, Sahai A, Kanwar YS. Immunomodulatory effects of HMG-CoA reductase inhibitors. Arch Immunol Ther Exp. 2003;51:139–48.
150.
go back to reference Sarkey JP, Richards MP, Stubbs EB Jr. Lovastatin attenuates nerve injury in an animal model of Guillain-Barre syndrome. J Neurochem. 2007;100:1265–77.PubMedCrossRef Sarkey JP, Richards MP, Stubbs EB Jr. Lovastatin attenuates nerve injury in an animal model of Guillain-Barre syndrome. J Neurochem. 2007;100:1265–77.PubMedCrossRef
151.
go back to reference Langert KA, Pervan CL, Stubbs EB Jr. Novel role of Cdc42 and RalA GTPases in TNF-alpha mediated secretion of CCL2. Small GTPases. 2014;5. Langert KA, Pervan CL, Stubbs EB Jr. Novel role of Cdc42 and RalA GTPases in TNF-alpha mediated secretion of CCL2. Small GTPases. 2014;5.
152.
go back to reference Takeuchi S, Kawashima S, Rikitake Y, Ueyama T, Inoue N, Hirata K, Yokoyama M. Cerivastatin suppresses lipopolysaccharide-induced ICAM-1 expression through inhibition of rho GTPase in BAEC. Biochem Biophys Res Commun. 2000;269:97–102.PubMedCrossRef Takeuchi S, Kawashima S, Rikitake Y, Ueyama T, Inoue N, Hirata K, Yokoyama M. Cerivastatin suppresses lipopolysaccharide-induced ICAM-1 expression through inhibition of rho GTPase in BAEC. Biochem Biophys Res Commun. 2000;269:97–102.PubMedCrossRef
153.
go back to reference Lin R, Liu J, Peng N, Gan W, Wang W, Han C, Ding C. Lovastatin reduces apoptosis and downregulates the CD40 expression induced by TNF-alpha in cerebral vascular endothelial cells. Curr Neurovasc Res. 2006;3:41–7.PubMedCrossRef Lin R, Liu J, Peng N, Gan W, Wang W, Han C, Ding C. Lovastatin reduces apoptosis and downregulates the CD40 expression induced by TNF-alpha in cerebral vascular endothelial cells. Curr Neurovasc Res. 2006;3:41–7.PubMedCrossRef
154.
go back to reference Stach K, Nguyen XD, Lang S, Elmas E, Weiss C, Borggrefe M, Fischer J, Kalsch T. Simvastatin and atorvastatin attenuate VCAM-1 and uPAR expression on human endothelial cells and platelet surface expression of CD40 ligand. Cardiol J. 2012;19:20–8.PubMedCrossRef Stach K, Nguyen XD, Lang S, Elmas E, Weiss C, Borggrefe M, Fischer J, Kalsch T. Simvastatin and atorvastatin attenuate VCAM-1 and uPAR expression on human endothelial cells and platelet surface expression of CD40 ligand. Cardiol J. 2012;19:20–8.PubMedCrossRef
155.
go back to reference Ho MH, Chiang CP, Liu YF, Kuo MY, Lin SK, Lai JY, Lee BS. Highly efficient release of lovastatin from poly(lactic-co-glycolic acid) nanoparticles enhances bone repair in rats. J Orthop Res. 2011;29:1504–10.PubMedCrossRef Ho MH, Chiang CP, Liu YF, Kuo MY, Lin SK, Lai JY, Lee BS. Highly efficient release of lovastatin from poly(lactic-co-glycolic acid) nanoparticles enhances bone repair in rats. J Orthop Res. 2011;29:1504–10.PubMedCrossRef
156.
go back to reference Langert KA, Goshu B, Stubbs EB Jr. Attenuation of experimental autoimmune neuritis with locally administered lovastatin-encapsulating poly(lactic-co-glycolic) acid nanoparticles. J Neurochem. 2017;140:334–46.PubMedCrossRef Langert KA, Goshu B, Stubbs EB Jr. Attenuation of experimental autoimmune neuritis with locally administered lovastatin-encapsulating poly(lactic-co-glycolic) acid nanoparticles. J Neurochem. 2017;140:334–46.PubMedCrossRef
157.
go back to reference Baumer P, Weiler M, Ruetters M, Staub F, Dombert T, Heiland S, Bendszus M, Pham M. MR neurography in ulnar neuropathy as surrogate parameter for the presence of disseminated neuropathy. PLoS One. 2012;7:e49742.PubMedPubMedCentralCrossRef Baumer P, Weiler M, Ruetters M, Staub F, Dombert T, Heiland S, Bendszus M, Pham M. MR neurography in ulnar neuropathy as surrogate parameter for the presence of disseminated neuropathy. PLoS One. 2012;7:e49742.PubMedPubMedCentralCrossRef
158.
go back to reference Kollmer J, Bendszus M, Pham M. MR Neurography: diagnostic imaging in the PNS. Clin Neuroradiol. 2015;25(Suppl 2):283–9.PubMedCrossRef Kollmer J, Bendszus M, Pham M. MR Neurography: diagnostic imaging in the PNS. Clin Neuroradiol. 2015;25(Suppl 2):283–9.PubMedCrossRef
159.
go back to reference Stoll G, Bendszus M. Imaging of inflammation in the peripheral and central nervous system by magnetic resonance imaging. Neuroscience. 2009;158:1151–60.PubMedCrossRef Stoll G, Bendszus M. Imaging of inflammation in the peripheral and central nervous system by magnetic resonance imaging. Neuroscience. 2009;158:1151–60.PubMedCrossRef
160.
go back to reference Anderson SA, Shukaliak-Quandt J, Jordan EK, Arbab AS, Martin R, McFarland H, Frank JA. Magnetic resonance imaging of labeled T-cells in a mouse model of multiple sclerosis. Ann Neurol. 2004;55:654–9.PubMedCrossRef Anderson SA, Shukaliak-Quandt J, Jordan EK, Arbab AS, Martin R, McFarland H, Frank JA. Magnetic resonance imaging of labeled T-cells in a mouse model of multiple sclerosis. Ann Neurol. 2004;55:654–9.PubMedCrossRef
161.
go back to reference Rausch M, Hiestand P, Foster CA, Baumann DR, Cannet C, Rudin M. Predictability of FTY720 efficacy in experimental autoimmune encephalomyelitis by in vivo macrophage tracking: clinical implications for ultrasmall superparamagnetic iron oxide-enhanced magnetic resonance imaging. J Magn Reson Imaging. 2004;20:16–24.PubMedCrossRef Rausch M, Hiestand P, Foster CA, Baumann DR, Cannet C, Rudin M. Predictability of FTY720 efficacy in experimental autoimmune encephalomyelitis by in vivo macrophage tracking: clinical implications for ultrasmall superparamagnetic iron oxide-enhanced magnetic resonance imaging. J Magn Reson Imaging. 2004;20:16–24.PubMedCrossRef
162.
go back to reference Chen P, Piao X, Bonaldo P. Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol. 2015;130:605–18.PubMedCrossRef Chen P, Piao X, Bonaldo P. Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol. 2015;130:605–18.PubMedCrossRef
163.
go back to reference Stoll G, Wesemeier C, Gold R, Solymosi L, Toyka KV, Bendszus M. In vivo monitoring of macrophage infiltration in experimental autoimmune neuritis by magnetic resonance imaging. J Neuroimmunol. 2004;149:142–6.PubMedCrossRef Stoll G, Wesemeier C, Gold R, Solymosi L, Toyka KV, Bendszus M. In vivo monitoring of macrophage infiltration in experimental autoimmune neuritis by magnetic resonance imaging. J Neuroimmunol. 2004;149:142–6.PubMedCrossRef
164.
go back to reference Stoll G, Wessig C, Gold R, Bendszus M. Assessment of lesion evolution in experimental autoimmune neuritis by gadofluorine M-enhanced MR neurography. Exp Neurol. 2006;197:150–6.PubMedCrossRef Stoll G, Wessig C, Gold R, Bendszus M. Assessment of lesion evolution in experimental autoimmune neuritis by gadofluorine M-enhanced MR neurography. Exp Neurol. 2006;197:150–6.PubMedCrossRef
165.
go back to reference Kirschbaum K, Sonner JK, Zeller MW, Deumelandt K, Bode J, Sharma R, Kruwel T, Fischer M, Hoffmann A, Costa da Silva M, et al. In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis. Proc Natl Acad Sci U S A. 2016;113:13227–32.PubMedPubMedCentralCrossRef Kirschbaum K, Sonner JK, Zeller MW, Deumelandt K, Bode J, Sharma R, Kruwel T, Fischer M, Hoffmann A, Costa da Silva M, et al. In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis. Proc Natl Acad Sci U S A. 2016;113:13227–32.PubMedPubMedCentralCrossRef
166.
go back to reference Willison HJ, Jacobs BC, van Doorn PA. Guillain-Barre syndrome. Lancet. 2016;388:717–27. Willison HJ, Jacobs BC, van Doorn PA. Guillain-Barre syndrome. Lancet. 2016;388:717–27.
167.
go back to reference Kleyman I, Brannagan TH 3rd. Treatment of chronic inflammatory demyelinating polyneuropathy. Curr Neurol Neurosci Rep. 2015;15:47.PubMedCrossRef Kleyman I, Brannagan TH 3rd. Treatment of chronic inflammatory demyelinating polyneuropathy. Curr Neurol Neurosci Rep. 2015;15:47.PubMedCrossRef
168.
go back to reference Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, Thornton R, Shei GJ, Card D, Keohane C, et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296:346–9.PubMedCrossRef Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, Thornton R, Shei GJ, Card D, Keohane C, et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296:346–9.PubMedCrossRef
169.
go back to reference Miron VE, Jung CG, Kim HJ, Kennedy TE, Soliven B, Antel JP. FTY720 modulates human oligodendrocyte progenitor process extension and survival. Ann Neurol. 2008;63:61–71.PubMedCrossRef Miron VE, Jung CG, Kim HJ, Kennedy TE, Soliven B, Antel JP. FTY720 modulates human oligodendrocyte progenitor process extension and survival. Ann Neurol. 2008;63:61–71.PubMedCrossRef
170.
go back to reference Balatoni B, Storch MK, Swoboda EM, Schonborn V, Koziel A, Lambrou GN, Hiestand PC, Weissert R, Foster CA. FTY720 sustains and restores neuronal function in the DA rat model of MOG-induced experimental autoimmune encephalomyelitis. Brain Res Bull. 2007;74:307–16.PubMedCrossRef Balatoni B, Storch MK, Swoboda EM, Schonborn V, Koziel A, Lambrou GN, Hiestand PC, Weissert R, Foster CA. FTY720 sustains and restores neuronal function in the DA rat model of MOG-induced experimental autoimmune encephalomyelitis. Brain Res Bull. 2007;74:307–16.PubMedCrossRef
171.
go back to reference Fujino M, Funeshima N, Kitazawa Y, Kimura H, Amemiya H, Suzuki S, Li XK. Amelioration of experimental autoimmune encephalomyelitis in Lewis rats by FTY720 treatment. J Pharmacol Exp Ther. 2003;305:70–7.PubMedCrossRef Fujino M, Funeshima N, Kitazawa Y, Kimura H, Amemiya H, Suzuki S, Li XK. Amelioration of experimental autoimmune encephalomyelitis in Lewis rats by FTY720 treatment. J Pharmacol Exp Ther. 2003;305:70–7.PubMedCrossRef
172.
go back to reference Webb M, Tham CS, Lin FF, Lariosa-Willingham K, Yu N, Hale J, Mandala S, Chun J, Rao TS. Sphingosine 1-phosphate receptor agonists attenuate relapsing-remitting experimental autoimmune encephalitis in SJL mice. J Neuroimmunol. 2004;153:108–21.PubMedCrossRef Webb M, Tham CS, Lin FF, Lariosa-Willingham K, Yu N, Hale J, Mandala S, Chun J, Rao TS. Sphingosine 1-phosphate receptor agonists attenuate relapsing-remitting experimental autoimmune encephalitis in SJL mice. J Neuroimmunol. 2004;153:108–21.PubMedCrossRef
173.
go back to reference Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, Calabresi P, Selmaj K, Agoropoulou C, Leyk M, Zhang-Auberson L, Burtin P. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362:387–401.PubMedCrossRef Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, Calabresi P, Selmaj K, Agoropoulou C, Leyk M, Zhang-Auberson L, Burtin P. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362:387–401.PubMedCrossRef
174.
go back to reference Khatri BO. Fingolimod in the treatment of relapsing-remitting multiple sclerosis: long-term experience and an update on the clinical evidence. Ther Adv Neurol Disord. 2016;9:130–47.PubMedPubMedCentralCrossRef Khatri BO. Fingolimod in the treatment of relapsing-remitting multiple sclerosis: long-term experience and an update on the clinical evidence. Ther Adv Neurol Disord. 2016;9:130–47.PubMedPubMedCentralCrossRef
175.
go back to reference Ambrosius B, Pitarokoili K, Schrewe L, Pedreiturria X, Motte J, Gold R. Fingolimod attenuates experimental autoimmune neuritis and contributes to Schwann cell-mediated axonal protection. J Neuroinflammation. 2017;14:92.PubMedPubMedCentralCrossRef Ambrosius B, Pitarokoili K, Schrewe L, Pedreiturria X, Motte J, Gold R. Fingolimod attenuates experimental autoimmune neuritis and contributes to Schwann cell-mediated axonal protection. J Neuroinflammation. 2017;14:92.PubMedPubMedCentralCrossRef
176.
go back to reference Heinen A, Beyer F, Tzekova N, Hartung HP, Kury P. Fingolimod induces the transition to a nerve regeneration promoting Schwann cell phenotype. Exp Neurol. 2015;271:25–35.PubMedCrossRef Heinen A, Beyer F, Tzekova N, Hartung HP, Kury P. Fingolimod induces the transition to a nerve regeneration promoting Schwann cell phenotype. Exp Neurol. 2015;271:25–35.PubMedCrossRef
177.
go back to reference Thomas K, Sehr T, Proschmann U, Rodriguez-Leal FA, Haase R, Ziemssen T. Fingolimod additionally acts as immunomodulator focused on the innate immune system beyond its prominent effects on lymphocyte recirculation. J Neuroinflammation. 2017;14:41.PubMedPubMedCentralCrossRef Thomas K, Sehr T, Proschmann U, Rodriguez-Leal FA, Haase R, Ziemssen T. Fingolimod additionally acts as immunomodulator focused on the innate immune system beyond its prominent effects on lymphocyte recirculation. J Neuroinflammation. 2017;14:41.PubMedPubMedCentralCrossRef
178.
go back to reference Hartung H-P, Dalakas M, Merkies I, Latov N, Léger J-M, Nobile-Orazio E, Sobue G, Genge A, Merschhemke MB, Marie EC, et al. Oral Fingolimod in chronic inflammatory Demyelinating Polyradiculoneuropathy (FORCIDP): results from a phase III randomized placebo-controlled trial (S27.002). Neurology. 2017;88. Hartung H-P, Dalakas M, Merkies I, Latov N, Léger J-M, Nobile-Orazio E, Sobue G, Genge A, Merschhemke MB, Marie EC, et al. Oral Fingolimod in chronic inflammatory Demyelinating Polyradiculoneuropathy (FORCIDP): results from a phase III randomized placebo-controlled trial (S27.002). Neurology. 2017;88.
179.
go back to reference Binder M, Otto F, Mertelsmann R, Veelken H, Trepel M. The epitope recognized by rituximab. Blood. 2006;108:1975–8.PubMedCrossRef Binder M, Otto F, Mertelsmann R, Veelken H, Trepel M. The epitope recognized by rituximab. Blood. 2006;108:1975–8.PubMedCrossRef
180.
go back to reference Barun B, Bar-Or A. Treatment of multiple sclerosis with anti-CD20 antibodies. Clin Immunol. 2011;142:31–37. Barun B, Bar-Or A. Treatment of multiple sclerosis with anti-CD20 antibodies. Clin Immunol. 2011;142:31–37.
181.
go back to reference Vidal L, Gafter-Gvili A, Salles G, Dreyling MH, Ghielmini M, Hsu Schmitz SF, Pettengell R, Witzens-Harig M, Shpilberg O. Rituximab maintenance for the treatment of patients with follicular lymphoma: an updated systematic review and meta-analysis of randomized trials. J Natl Cancer Inst. 2011;103:1799–806.PubMedCrossRef Vidal L, Gafter-Gvili A, Salles G, Dreyling MH, Ghielmini M, Hsu Schmitz SF, Pettengell R, Witzens-Harig M, Shpilberg O. Rituximab maintenance for the treatment of patients with follicular lymphoma: an updated systematic review and meta-analysis of randomized trials. J Natl Cancer Inst. 2011;103:1799–806.PubMedCrossRef
182.
go back to reference Sorensen PS, Blinkenberg M. The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects. Ther Adv Neurol Disord. 2016;9:44–52.PubMedPubMedCentralCrossRef Sorensen PS, Blinkenberg M. The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects. Ther Adv Neurol Disord. 2016;9:44–52.PubMedPubMedCentralCrossRef
183.
go back to reference Kappos L, Li D, Calabresi PA, O’Connor P, Bar-Or A, Barkhof F, Yin M, Leppert D, Glanzman R, Tinbergen J, Hauser SL. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet. 2011;378:1779–87.PubMedCrossRef Kappos L, Li D, Calabresi PA, O’Connor P, Bar-Or A, Barkhof F, Yin M, Leppert D, Glanzman R, Tinbergen J, Hauser SL. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet. 2011;378:1779–87.PubMedCrossRef
184.
go back to reference Hauser SL, Bar-Or A, Comi G, Giovannoni G, Hartung HP, Hemmer B, Lublin F, Montalban X, Rammohan KW, Selmaj K, et al. Ocrelizumab versus interferon Beta-1a in relapsing multiple sclerosis. N Engl J Med. 2016;376:221–34. Hauser SL, Bar-Or A, Comi G, Giovannoni G, Hartung HP, Hemmer B, Lublin F, Montalban X, Rammohan KW, Selmaj K, et al. Ocrelizumab versus interferon Beta-1a in relapsing multiple sclerosis. N Engl J Med. 2016;376:221–34.
185.
go back to reference Montalban X, Hauser SL, Kappos L, Arnold DL, Bar-Or A, Comi G, de Seze J, Giovannoni G, Hartung HP, Hemmer B, et al. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. 2017;376:209–20.PubMedCrossRef Montalban X, Hauser SL, Kappos L, Arnold DL, Bar-Or A, Comi G, de Seze J, Giovannoni G, Hartung HP, Hemmer B, et al. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. 2017;376:209–20.PubMedCrossRef
186.
go back to reference Sorensen PS, Lisby S, Grove R, Derosier F, Shackelford S, Havrdova E, Drulovic J, Filippi M. Safety and efficacy of ofatumumab in relapsing-remitting multiple sclerosis: a phase 2 study. Neurology. 2014;82:573–81.PubMedCrossRef Sorensen PS, Lisby S, Grove R, Derosier F, Shackelford S, Havrdova E, Drulovic J, Filippi M. Safety and efficacy of ofatumumab in relapsing-remitting multiple sclerosis: a phase 2 study. Neurology. 2014;82:573–81.PubMedCrossRef
187.
go back to reference Meyer Zu Horste G, Mausberg AK, Korth C, Stuve O, Kieseier BC. Quinpramine-a promising compound for treating immune-mediated demyelination of the nervous system. Drug News Perspect. 2010;23:287–94.PubMedCrossRef Meyer Zu Horste G, Mausberg AK, Korth C, Stuve O, Kieseier BC. Quinpramine-a promising compound for treating immune-mediated demyelination of the nervous system. Drug News Perspect. 2010;23:287–94.PubMedCrossRef
188.
go back to reference Klingenstein R, Lober S, Kujala P, Godsave S, Leliveld SR, Gmeiner P, Peters PJ, Korth C. Tricyclic antidepressants, quinacrine and a novel, synthetic chimera thereof clear prions by destabilizing detergent-resistant membrane compartments. J Neurochem. 2006;98:748–59.PubMedCrossRef Klingenstein R, Lober S, Kujala P, Godsave S, Leliveld SR, Gmeiner P, Peters PJ, Korth C. Tricyclic antidepressants, quinacrine and a novel, synthetic chimera thereof clear prions by destabilizing detergent-resistant membrane compartments. J Neurochem. 2006;98:748–59.PubMedCrossRef
189.
go back to reference Singh MP, Meyer zu Horste G, Hu W, Mausberg AK, Cravens PD, Eagar T, Lober S, Klingenstein R, Gmeiner P, Korth C, et al. Quinpramine is a novel compound effective in ameliorating brain autoimmune disease. Exp Neurol. 2009;215:397–400.PubMedCrossRef Singh MP, Meyer zu Horste G, Hu W, Mausberg AK, Cravens PD, Eagar T, Lober S, Klingenstein R, Gmeiner P, Korth C, et al. Quinpramine is a novel compound effective in ameliorating brain autoimmune disease. Exp Neurol. 2009;215:397–400.PubMedCrossRef
190.
go back to reference Meyer Zu Horste G, Mausberg AK, Muller JI, Lehmann HC, Lober S, Gmeiner P, Hartung HP, Stuve O, Korth C, Kieseier BC. Quinpramine ameliorates rat experimental autoimmune neuritis and redistributes MHC class II molecules. PLoS One. 2011;6:e21223.PubMedPubMedCentralCrossRef Meyer Zu Horste G, Mausberg AK, Muller JI, Lehmann HC, Lober S, Gmeiner P, Hartung HP, Stuve O, Korth C, Kieseier BC. Quinpramine ameliorates rat experimental autoimmune neuritis and redistributes MHC class II molecules. PLoS One. 2011;6:e21223.PubMedPubMedCentralCrossRef
191.
go back to reference Milo R. Therapeutic strategies targeting B-cells in multiple sclerosis. Autoimmun Rev. 2016;15:714–8.PubMedCrossRef Milo R. Therapeutic strategies targeting B-cells in multiple sclerosis. Autoimmun Rev. 2016;15:714–8.PubMedCrossRef
192.
go back to reference Marsh EA, Hirst CL, Llewelyn JG, Cossburn MD, Reilly MM, Krishnan A, Doran M, Ryan AM, Coles AJ, Jones JL, Robertson NP. Alemtuzumab in the treatment of IVIG-dependent chronic inflammatory demyelinating polyneuropathy. J Neurol. 2010;257:913–9.PubMedCrossRef Marsh EA, Hirst CL, Llewelyn JG, Cossburn MD, Reilly MM, Krishnan A, Doran M, Ryan AM, Coles AJ, Jones JL, Robertson NP. Alemtuzumab in the treatment of IVIG-dependent chronic inflammatory demyelinating polyneuropathy. J Neurol. 2010;257:913–9.PubMedCrossRef
193.
go back to reference Hirst C, Raasch S, Llewelyn G, Robertson N. Remission of chronic inflammatory demyelinating polyneuropathy after alemtuzumab (Campath 1H). J Neurol Neurosurg Psychiatry. 2006;77:800–2.PubMedPubMedCentralCrossRef Hirst C, Raasch S, Llewelyn G, Robertson N. Remission of chronic inflammatory demyelinating polyneuropathy after alemtuzumab (Campath 1H). J Neurol Neurosurg Psychiatry. 2006;77:800–2.PubMedPubMedCentralCrossRef
194.
go back to reference Tzachanis D, Hamdan A, Uhlmann EJ, Joyce RM. Successful treatment of refractory Guillain-Barre syndrome with alemtuzumab in a patient with chronic lymphocytic leukemia. Acta Haematol. 2014;132:240–3.PubMedCrossRef Tzachanis D, Hamdan A, Uhlmann EJ, Joyce RM. Successful treatment of refractory Guillain-Barre syndrome with alemtuzumab in a patient with chronic lymphocytic leukemia. Acta Haematol. 2014;132:240–3.PubMedCrossRef
195.
go back to reference Thomas K, Eisele J, Rodriguez-Leal FA, Hainke U, Ziemssen T. Acute effects of alemtuzumab infusion in patients with active relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm. 2016;3:e228.PubMedPubMedCentralCrossRef Thomas K, Eisele J, Rodriguez-Leal FA, Hainke U, Ziemssen T. Acute effects of alemtuzumab infusion in patients with active relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm. 2016;3:e228.PubMedPubMedCentralCrossRef
196.
go back to reference Moreland LW, Baumgartner SW, Schiff MH, Tindall EA, Fleischmann RM, Weaver AL, Ettlinger RE, Cohen S, Koopman WJ, Mohler K, et al. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N Engl J Med. 1997;337:141–7.PubMedCrossRef Moreland LW, Baumgartner SW, Schiff MH, Tindall EA, Fleischmann RM, Weaver AL, Ettlinger RE, Cohen S, Koopman WJ, Mohler K, et al. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N Engl J Med. 1997;337:141–7.PubMedCrossRef
197.
go back to reference Chin RL, Sherman WH, Sander HW, Hays AP, Latov N. Etanercept (Enbrel) therapy for chronic inflammatory demyelinating polyneuropathy. J Neurol Sci. 2003;210:19–21.PubMedCrossRef Chin RL, Sherman WH, Sander HW, Hays AP, Latov N. Etanercept (Enbrel) therapy for chronic inflammatory demyelinating polyneuropathy. J Neurol Sci. 2003;210:19–21.PubMedCrossRef
198.
go back to reference Andreadou E, Kemanetzoglou E, Brokalaki C, Evangelopoulos ME, Kilidireas C, Rombos A, Stamboulis E. Demyelinating disease following anti-TNFa treatment: a causal or coincidental association? Report of four cases and review of the literature. Case Rep Neurol Med. 2013;2013:671935.PubMedPubMedCentral Andreadou E, Kemanetzoglou E, Brokalaki C, Evangelopoulos ME, Kilidireas C, Rombos A, Stamboulis E. Demyelinating disease following anti-TNFa treatment: a causal or coincidental association? Report of four cases and review of the literature. Case Rep Neurol Med. 2013;2013:671935.PubMedPubMedCentral
199.
go back to reference Wanschitz J, Maier H, Lassmann H, Budka H, Berger T. Distinct time pattern of complement activation and cytotoxic T cell response in Guillain-Barre syndrome. Brain. 2003;126:2034–42.PubMedCrossRef Wanschitz J, Maier H, Lassmann H, Budka H, Berger T. Distinct time pattern of complement activation and cytotoxic T cell response in Guillain-Barre syndrome. Brain. 2003;126:2034–42.PubMedCrossRef
200.
go back to reference Susuki K, Rasband MN, Tohyama K, Koibuchi K, Okamoto S, Funakoshi K, Hirata K, Baba H, Yuki N. Anti-GM1 antibodies cause complement-mediated disruption of sodium channel clusters in peripheral motor nerve fibers. J Neurosci. 2007;27:3956–67.PubMedCrossRef Susuki K, Rasband MN, Tohyama K, Koibuchi K, Okamoto S, Funakoshi K, Hirata K, Baba H, Yuki N. Anti-GM1 antibodies cause complement-mediated disruption of sodium channel clusters in peripheral motor nerve fibers. J Neurosci. 2007;27:3956–67.PubMedCrossRef
201.
go back to reference McGonigal R, Rowan EG, Greenshields KN, Halstead SK, Humphreys PD, Rother RP, Furukawa K, Willison HJ. Anti-GD1a antibodies activate complement and calpain to injure distal motor nodes of Ranvier in mice. Brain. 2010;133:1944–60.PubMedCrossRef McGonigal R, Rowan EG, Greenshields KN, Halstead SK, Humphreys PD, Rother RP, Furukawa K, Willison HJ. Anti-GD1a antibodies activate complement and calpain to injure distal motor nodes of Ranvier in mice. Brain. 2010;133:1944–60.PubMedCrossRef
202.
go back to reference Willison HJ, Halstead SK, Beveridge E, Zitman FM, Greenshields KN, Morgan BP, Plomp JJ. The role of complement and complement regulators in mediating motor nerve terminal injury in murine models of Guillain-Barre syndrome. J Neuroimmunol. 2008;201-202:172–82.PubMedCrossRef Willison HJ, Halstead SK, Beveridge E, Zitman FM, Greenshields KN, Morgan BP, Plomp JJ. The role of complement and complement regulators in mediating motor nerve terminal injury in murine models of Guillain-Barre syndrome. J Neuroimmunol. 2008;201-202:172–82.PubMedCrossRef
203.
go back to reference Jung S, Toyka KV, Hartung H-P. Soluble complement receptor type 1 inhibits experimental autoimmune neuritis in Lewis rats. Neurosci Lett. 1995;200:167–70.PubMedCrossRef Jung S, Toyka KV, Hartung H-P. Soluble complement receptor type 1 inhibits experimental autoimmune neuritis in Lewis rats. Neurosci Lett. 1995;200:167–70.PubMedCrossRef
204.
go back to reference Vriesendorp FJ, Flynn RE, Malone MR, Pappolla MA. Systemic complement depletion reduces inflammation and demyelination in adoptive transfer experimental allergic neuritis. Acta Neuropathol (Berl). 1998;95:297–301.CrossRef Vriesendorp FJ, Flynn RE, Malone MR, Pappolla MA. Systemic complement depletion reduces inflammation and demyelination in adoptive transfer experimental allergic neuritis. Acta Neuropathol (Berl). 1998;95:297–301.CrossRef
205.
go back to reference Halstead SK, Humphreys PD, Goodfellow JA, Wagner ER, Smith RA, Willison HJ. Complement inhibition abrogates nerve terminal injury in Miller Fisher syndrome. Ann Neurol. 2005;58:203–10.PubMedCrossRef Halstead SK, Humphreys PD, Goodfellow JA, Wagner ER, Smith RA, Willison HJ. Complement inhibition abrogates nerve terminal injury in Miller Fisher syndrome. Ann Neurol. 2005;58:203–10.PubMedCrossRef
206.
go back to reference Tran GT, Hodgkinson SJ, Carter NM, Killingsworth M, Nomura M, Verma ND, Plain KM, Boyd R, Hall BM. Membrane attack complex of complement is not essential for immune mediated demyelination in experimental autoimmune neuritis. J Neuroimmunol. 2010;229:98–106.PubMedCrossRef Tran GT, Hodgkinson SJ, Carter NM, Killingsworth M, Nomura M, Verma ND, Plain KM, Boyd R, Hall BM. Membrane attack complex of complement is not essential for immune mediated demyelination in experimental autoimmune neuritis. J Neuroimmunol. 2010;229:98–106.PubMedCrossRef
207.
go back to reference Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, Dilthey A, Su Z, Freeman C, Hunt SE, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476:214–9.PubMedPubMedCentralCrossRef Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, Dilthey A, Su Z, Freeman C, Hunt SE, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476:214–9.PubMedPubMedCentralCrossRef
208.
go back to reference Maurer M, Gold R. Animal models of immune-mediated neuropathies. Curr Opin Neurol. 2002;15:617–22.PubMedCrossRef Maurer M, Gold R. Animal models of immune-mediated neuropathies. Curr Opin Neurol. 2002;15:617–22.PubMedCrossRef
209.
go back to reference Deretzi G, Zou LP, Pelidou SH, Nennesmo I, Levi M, Wahren B, Mix E, Zhu J. Nasal administration of recombinant rat IL-4 ameliorates ongoing experimental autoimmune neuritis and inhibits demyelination. J Autoimmun. 1999;12:81–9.PubMedCrossRef Deretzi G, Zou LP, Pelidou SH, Nennesmo I, Levi M, Wahren B, Mix E, Zhu J. Nasal administration of recombinant rat IL-4 ameliorates ongoing experimental autoimmune neuritis and inhibits demyelination. J Autoimmun. 1999;12:81–9.PubMedCrossRef
210.
go back to reference Miyamoto K, Oka N, Kawasaki T, Satoi H, Matsuo A, Akiguchi I. The action mechanism of cyclooxygenase-2 inhibitor for treatment of experimental allergic neuritis. Muscle Nerve. 1999;22:1704–9.PubMedCrossRef Miyamoto K, Oka N, Kawasaki T, Satoi H, Matsuo A, Akiguchi I. The action mechanism of cyclooxygenase-2 inhibitor for treatment of experimental allergic neuritis. Muscle Nerve. 1999;22:1704–9.PubMedCrossRef
211.
go back to reference Abbas N, Zou LP, Pelidou SH, Winblad B, Zhu J. Protective effect of Rolipram in experimental autoimmune neuritis: protection is associated with down-regulation of IFN-gamma and inflammatory chemokines as well as up-regulation of IL-4 in peripheral nervous system. Autoimmunity. 2000;32:93–9.PubMedCrossRef Abbas N, Zou LP, Pelidou SH, Winblad B, Zhu J. Protective effect of Rolipram in experimental autoimmune neuritis: protection is associated with down-regulation of IFN-gamma and inflammatory chemokines as well as up-regulation of IL-4 in peripheral nervous system. Autoimmunity. 2000;32:93–9.PubMedCrossRef
212.
go back to reference Hoffman PM, Powers JM, Weise MJ, Brostoff SW. Experimental allergic neuritis. I. Rat strain differences in the response to bovine myelin antigens. Brain Res. 1980;195:355–62.PubMedCrossRef Hoffman PM, Powers JM, Weise MJ, Brostoff SW. Experimental allergic neuritis. I. Rat strain differences in the response to bovine myelin antigens. Brain Res. 1980;195:355–62.PubMedCrossRef
213.
go back to reference Miletic H, Utermohlen O, Wedekind C, Hermann M, Stenzel W, Lassmann H, Schluter D, Deckert M. P0(106-125) is a neuritogenic epitope of the peripheral myelin protein P0 and induces autoimmune neuritis in C57BL/6 mice. J Neuropathol Exp Neurol. 2005;64:66–73.PubMedCrossRef Miletic H, Utermohlen O, Wedekind C, Hermann M, Stenzel W, Lassmann H, Schluter D, Deckert M. P0(106-125) is a neuritogenic epitope of the peripheral myelin protein P0 and induces autoimmune neuritis in C57BL/6 mice. J Neuropathol Exp Neurol. 2005;64:66–73.PubMedCrossRef
214.
go back to reference Zou LP, Ljunggren HG, Levi M, Nennesmo I, Wahren B, Mix E, Winblad B, Schalling M, Zhu J. P0 protein peptide 180-199 together with pertussis toxin induces experimental autoimmune neuritis in resistant C57BL/6 mice. J Neurosci Res. 2000;62:717–21.PubMedCrossRef Zou LP, Ljunggren HG, Levi M, Nennesmo I, Wahren B, Mix E, Winblad B, Schalling M, Zhu J. P0 protein peptide 180-199 together with pertussis toxin induces experimental autoimmune neuritis in resistant C57BL/6 mice. J Neurosci Res. 2000;62:717–21.PubMedCrossRef
215.
go back to reference Taylor WA, Hughes RA. Experimental allergic neuritis induced in SJL mice by bovine P2. J Neuroimmunol. 1985;8:153–7.PubMedCrossRef Taylor WA, Hughes RA. Experimental allergic neuritis induced in SJL mice by bovine P2. J Neuroimmunol. 1985;8:153–7.PubMedCrossRef
216.
go back to reference Calida DM, Kremlev SG, Fujioka T, Hilliard B, Ventura E, Constantinescu CS, Lavi E, Rostami A. Experimental allergic neuritis in the SJL/J mouse: induction of severe and reproducible disease with bovine peripheral nerve myelin and pertussis toxin with or without interleukin-12. J Neuroimmunol. 2000;107:1–7.PubMedCrossRef Calida DM, Kremlev SG, Fujioka T, Hilliard B, Ventura E, Constantinescu CS, Lavi E, Rostami A. Experimental allergic neuritis in the SJL/J mouse: induction of severe and reproducible disease with bovine peripheral nerve myelin and pertussis toxin with or without interleukin-12. J Neuroimmunol. 2000;107:1–7.PubMedCrossRef
217.
go back to reference Abromson-Leeman S, Bronson R, Dorf ME. Experimental autoimmune peripheral neuritis induced in BALB/c mice by myelin basic protein-specific T cell clones. J Exp Med. 1995;182:587–92.PubMedCrossRef Abromson-Leeman S, Bronson R, Dorf ME. Experimental autoimmune peripheral neuritis induced in BALB/c mice by myelin basic protein-specific T cell clones. J Exp Med. 1995;182:587–92.PubMedCrossRef
218.
go back to reference Snyder DH, Stone SH, Raine CS. Attempts to induce chronic experimental allergic neuritis in strain 13 and Hartley guinea pigs. J Neuropathol Exp Neurol. 1977;36:488–98.PubMedCrossRef Snyder DH, Stone SH, Raine CS. Attempts to induce chronic experimental allergic neuritis in strain 13 and Hartley guinea pigs. J Neuropathol Exp Neurol. 1977;36:488–98.PubMedCrossRef
219.
go back to reference Eylar EH, Toro-Goyco E, Kessler MJ, Szymanska I. Induction of allergic neuritis in rhesus monkeys. J Neuroimmunol. 1982;3:91–8.PubMedCrossRef Eylar EH, Toro-Goyco E, Kessler MJ, Szymanska I. Induction of allergic neuritis in rhesus monkeys. J Neuroimmunol. 1982;3:91–8.PubMedCrossRef
Metadata
Title
Novel pathomechanisms in inflammatory neuropathies
Authors
David Schafflick
Bernd C. Kieseier
Heinz Wiendl
Gerd Meyer zu Horste
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2017
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-1001-8

Other articles of this Issue 1/2017

Journal of Neuroinflammation 1/2017 Go to the issue