Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2019

Open Access 01-12-2019 | Filgrastim | Research

Anti-MOG autoantibodies pathogenicity in children and macaques demyelinating diseases

Authors: Che Serguera, Lev Stimmer, Claire-Maelle Fovet, Philippe Horellou, Vanessa Contreras, Nicolas Tchitchek, Julie Massonneau, Carole Leroy, Audrey Perrin, Julien Flament, Philippe Hantraye, Joanna Demilly, Romain Marignier, Pascale Chrétien, Bert‘t Hart, Jean Boutonnat, Clovis Adam, Roger Le-Grand, Kumaran Deiva

Published in: Journal of Neuroinflammation | Issue 1/2019

Login to get access

Abstract

Background

Autoantibodies against myelin oligodendrocyte glycoprotein (anti-MOG-Abs) occur in a majority of children with acquired demyelinating syndromes (ADS) and physiopathology is still under investigation. As cynomolgus macaques immunized with rhMOG, all develop an experimental autoimmune encephalomyelitis (EAE), we assessed relatedness between anti-MOG-Abs associated diseases in both species.

Methods

The study includes 27 children followed for ADS and nine macaques with rhMOG-induced EAE. MRI lesions, cytokines in blood, and CSF at onset of ADS or EAE, as well as histopathological features of brain lesions were compared.

Results

Twelve children with anti-MOG-Abs ADS (ADS MOG+) and nine macaques with EAE, presented increased IL-6 and G-CSF in the CSF, whereas no such signature was found in 15 ADS MOG−. Furthermore, IgG and C1q were associated to myelin and phagocytic cells in brains with EAE (n = 8) and in biopsies of ADS MOG+ (n = 2) but not ADS MOG− children (n = 1). Macaque brains also revealed prephagocytic lesions with IgG and C1q depositions but no leukocyte infiltration.

Conclusions

Children with ADS MOG+ and macaques with EAE induced with rhMOG, present a similar cytokine signature in the CSF and a comparable aspect of brain lesions indicating analogous pathophysiological processes. In EAE, prephagocytic lesions points at IgG as an initial effector of myelin attack. These results support the pertinence of modeling ADS MOG+ in non-human primates to apprehend the natural development of anti-MOG-associated disease, find markers of evolution, and above all explore the efficacy of targeted therapies to test primate-restricted molecules.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jarius S, Ruprecht K, Stellmann JP, Huss A, Ayzenberg I, Willing A, et al. MOG-IgG in primary and secondary chronic progressive multiple sclerosis: a multicenter study of 200 patients and review of the literature. J Neuroinflammation. 2018;15:88.CrossRef Jarius S, Ruprecht K, Stellmann JP, Huss A, Ayzenberg I, Willing A, et al. MOG-IgG in primary and secondary chronic progressive multiple sclerosis: a multicenter study of 200 patients and review of the literature. J Neuroinflammation. 2018;15:88.CrossRef
2.
go back to reference Hacohen Y, Wong YY, Lechner C, Jurynczyk M, Wright S, Konuskan B, et al. Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein antibody–associated disease. JAMA Neurol. 2018;75:478–87.CrossRef Hacohen Y, Wong YY, Lechner C, Jurynczyk M, Wright S, Konuskan B, et al. Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein antibody–associated disease. JAMA Neurol. 2018;75:478–87.CrossRef
3.
go back to reference t Hart BA, Gran B, Weissert R. EAE: imperfect but useful models of multiple sclerosis. Trends Mol Med. 2011;17:119–25. t Hart BA, Gran B, Weissert R. EAE: imperfect but useful models of multiple sclerosis. Trends Mol Med. 2011;17:119–25.
4.
go back to reference Spadaro M, Winklmeier S, Beltrán E, Macrini C, Höftberger R, Schuh E, et al. Pathogenicity of human antibodies against myelin oligodendrocyte glycoprotein. Ann Neurol. 2018;84:315–28.CrossRef Spadaro M, Winklmeier S, Beltrán E, Macrini C, Höftberger R, Schuh E, et al. Pathogenicity of human antibodies against myelin oligodendrocyte glycoprotein. Ann Neurol. 2018;84:315–28.CrossRef
5.
go back to reference Stimmer L, Fovet C-M, Serguera C. Experimental Models of autoimmune demyelinating diseases in nonhuman primates. Vet Pathol. 2018;55:27–41.CrossRef Stimmer L, Fovet C-M, Serguera C. Experimental Models of autoimmune demyelinating diseases in nonhuman primates. Vet Pathol. 2018;55:27–41.CrossRef
6.
go back to reference Charan J, Kantharia ND. How to calculate sample size in animal studies? J Pharmacol Pharmacother. 2013;4:303.CrossRef Charan J, Kantharia ND. How to calculate sample size in animal studies? J Pharmacol Pharmacother. 2013;4:303.CrossRef
7.
go back to reference Krupp LB, Tardieu M, Amato MP, Banwell B, Chitnis T, Dale RC, et al. International Pediatric Multiple Sclerosis Study Group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions. Mult Scler J. 2013;19:1261–7.CrossRef Krupp LB, Tardieu M, Amato MP, Banwell B, Chitnis T, Dale RC, et al. International Pediatric Multiple Sclerosis Study Group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions. Mult Scler J. 2013;19:1261–7.CrossRef
8.
go back to reference Horellou P, Wang M, Keo V, Chretien P, Serguera C, Waters P, et al. Increased interleukin-6 correlates with myelin oligodendrocyte glycoprotein antibodies in pediatric monophasic demyelinating diseases and multiple sclerosis. J Neuroimmunol. 2015;289:1–7.CrossRef Horellou P, Wang M, Keo V, Chretien P, Serguera C, Waters P, et al. Increased interleukin-6 correlates with myelin oligodendrocyte glycoprotein antibodies in pediatric monophasic demyelinating diseases and multiple sclerosis. J Neuroimmunol. 2015;289:1–7.CrossRef
9.
go back to reference Haanstra KG, Jagessar SA, Bauchet AL, Doussau M, Fovet CM, Heijmans N, et al. Induction of experimental autoimmune encephalomyelitis with recombinant human myelin oligodendrocyte glycoprotein in incomplete Freund’s adjuvant in three non-human primate species. J Neuroimmune Pharmacol. 2013;8:1251–64.CrossRef Haanstra KG, Jagessar SA, Bauchet AL, Doussau M, Fovet CM, Heijmans N, et al. Induction of experimental autoimmune encephalomyelitis with recombinant human myelin oligodendrocyte glycoprotein in incomplete Freund’s adjuvant in three non-human primate species. J Neuroimmune Pharmacol. 2013;8:1251–64.CrossRef
10.
go back to reference Kothur K, Wienholt L, Tantsis EM, Earl J, Bandodkar S, Prelog K, et al. B Cell, Th17, and Neutrophil Related Cerebrospinal Fluid Cytokine/Chemokines Are Elevated in MOG Antibody Associated Demyelination. PLOS ONE. 2016;11:e0149411.CrossRef Kothur K, Wienholt L, Tantsis EM, Earl J, Bandodkar S, Prelog K, et al. B Cell, Th17, and Neutrophil Related Cerebrospinal Fluid Cytokine/Chemokines Are Elevated in MOG Antibody Associated Demyelination. PLOS ONE. 2016;11:e0149411.CrossRef
11.
go back to reference Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. Handb Clin Neurol. 2017;145:263–83.CrossRef Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. Handb Clin Neurol. 2017;145:263–83.CrossRef
12.
go back to reference Barnett MH, Parratt JDE, Cho E-S, Prineas JW. Immunoglobulins and complement in postmortem multiple sclerosis tissue. Ann Neurol. 2009;65:32–46.CrossRef Barnett MH, Parratt JDE, Cho E-S, Prineas JW. Immunoglobulins and complement in postmortem multiple sclerosis tissue. Ann Neurol. 2009;65:32–46.CrossRef
13.
go back to reference Kaneko K, Sato DK, Nakashima I, Ogawa R, Akaishi T, Takai Y, et al. CSF cytokine profile in MOG-IgG+ neurological disease is similar to AQP4-IgG+ NMOSD but distinct from MS: a cross-sectional study and potential therapeutic implications. J Neurol Neurosurg Psychiatry. 2018;89:927–36.CrossRef Kaneko K, Sato DK, Nakashima I, Ogawa R, Akaishi T, Takai Y, et al. CSF cytokine profile in MOG-IgG+ neurological disease is similar to AQP4-IgG+ NMOSD but distinct from MS: a cross-sectional study and potential therapeutic implications. J Neurol Neurosurg Psychiatry. 2018;89:927–36.CrossRef
14.
go back to reference Matsushita T, Horikawa M, Iwata Y, Tedder TF. Regulatory B cells (B10 cells) and regulatory T cells have independent roles in controlling experimental autoimmune encephalomyelitis initiation and late-phase immunopathogenesis. J Immunol. 2010;185:2240–52.CrossRef Matsushita T, Horikawa M, Iwata Y, Tedder TF. Regulatory B cells (B10 cells) and regulatory T cells have independent roles in controlling experimental autoimmune encephalomyelitis initiation and late-phase immunopathogenesis. J Immunol. 2010;185:2240–52.CrossRef
15.
go back to reference Uzawa A, Mori M, Arai K, Sato Y, Hayakawa S, Masuda S, et al. Cytokine and chemokine profiles in neuromyelitis optica: significance of interleukin-6. Mult Scler J. 2010;16:1443–52.CrossRef Uzawa A, Mori M, Arai K, Sato Y, Hayakawa S, Masuda S, et al. Cytokine and chemokine profiles in neuromyelitis optica: significance of interleukin-6. Mult Scler J. 2010;16:1443–52.CrossRef
17.
go back to reference Jarius S, Metz I, König FB, Ruprecht K, Reindl M, Paul F, et al. Screening for MOG-IgG and 27 other anti-glial and anti-neuronal autoantibodies in “pattern II multiple sclerosis” and brain biopsy findings in a MOG-IgG-positive case. Mult Scler Houndmills Basingstoke Engl. 2016;22:1541–9.CrossRef Jarius S, Metz I, König FB, Ruprecht K, Reindl M, Paul F, et al. Screening for MOG-IgG and 27 other anti-glial and anti-neuronal autoantibodies in “pattern II multiple sclerosis” and brain biopsy findings in a MOG-IgG-positive case. Mult Scler Houndmills Basingstoke Engl. 2016;22:1541–9.CrossRef
18.
go back to reference König FB, Wildemann B, Nessler S, Zhou D, Hemmer B, Metz I, et al. Persistence of Immunopathological and Radiological Traits in Multiple Sclerosis. Arch Neurol. 2008;65:1527–32.CrossRef König FB, Wildemann B, Nessler S, Zhou D, Hemmer B, Metz I, et al. Persistence of Immunopathological and Radiological Traits in Multiple Sclerosis. Arch Neurol. 2008;65:1527–32.CrossRef
20.
go back to reference Spadaro M, Gerdes LA, Mayer MC, Ertl-Wagner B, Laurent S, Krumbholz M, et al. Histopathology and clinical course of MOG-antibody-associated encephalomyelitis. Ann Clin Transl Neurol. 2015;2:295–301.CrossRef Spadaro M, Gerdes LA, Mayer MC, Ertl-Wagner B, Laurent S, Krumbholz M, et al. Histopathology and clinical course of MOG-antibody-associated encephalomyelitis. Ann Clin Transl Neurol. 2015;2:295–301.CrossRef
21.
go back to reference Wang JJ, Jaunmuktane Z, Mummery C, Brandner S, Leary S, Trip SA. Inflammatory demyelination without astrocyte loss in MOG antibody–positive NMOSD. Neurology. 2016;87:229–31.CrossRef Wang JJ, Jaunmuktane Z, Mummery C, Brandner S, Leary S, Trip SA. Inflammatory demyelination without astrocyte loss in MOG antibody–positive NMOSD. Neurology. 2016;87:229–31.CrossRef
22.
go back to reference Hennes E-M, Baumann M, Schanda K, Anlar B, Bajer-Kornek B, Blaschek A, et al. Prognostic relevance of MOG antibodies in children with an acquired demyelinating syndrome. Neurology. 2017;89:900–8.CrossRef Hennes E-M, Baumann M, Schanda K, Anlar B, Bajer-Kornek B, Blaschek A, et al. Prognostic relevance of MOG antibodies in children with an acquired demyelinating syndrome. Neurology. 2017;89:900–8.CrossRef
23.
go back to reference Rose LM, Richards TL, Petersen R, Peterson J, Hruby S, Alvord EC. Remitting-relapsing EAE in nonhuman primates: a valid model of multiple sclerosis. Clin Immunol Immunopathol. 1991;59:1–15.CrossRef Rose LM, Richards TL, Petersen R, Peterson J, Hruby S, Alvord EC. Remitting-relapsing EAE in nonhuman primates: a valid model of multiple sclerosis. Clin Immunol Immunopathol. 1991;59:1–15.CrossRef
24.
go back to reference Genain CP, Cannella B, Hauser SL, Raine CS. Identification of autoantibodies associated with myelin damage in multiple sclerosis. Nat Med. 1999;5:170–5.CrossRef Genain CP, Cannella B, Hauser SL, Raine CS. Identification of autoantibodies associated with myelin damage in multiple sclerosis. Nat Med. 1999;5:170–5.CrossRef
25.
go back to reference Matsushita T, Tateishi T, Isobe N, Yonekawa T, Yamasaki R, Matsuse D, et al. Characteristic cerebrospinal fluid cytokine/chemokine profiles in neuromyelitis optica, relapsing remitting or primary progressive multiple sclerosis. PLOS ONE. 2013;8:e61835.CrossRef Matsushita T, Tateishi T, Isobe N, Yonekawa T, Yamasaki R, Matsuse D, et al. Characteristic cerebrospinal fluid cytokine/chemokine profiles in neuromyelitis optica, relapsing remitting or primary progressive multiple sclerosis. PLOS ONE. 2013;8:e61835.CrossRef
26.
go back to reference Fovet C-M, Stimmer L, Contreras V, Horellou P, Hubert A, Seddiki N, et al. Intradermal vaccination prevents anti-MOG autoimmune encephalomyelitis in macaques. EBioMedicine. 2019;47:492–505.CrossRef Fovet C-M, Stimmer L, Contreras V, Horellou P, Hubert A, Seddiki N, et al. Intradermal vaccination prevents anti-MOG autoimmune encephalomyelitis in macaques. EBioMedicine. 2019;47:492–505.CrossRef
Metadata
Title
Anti-MOG autoantibodies pathogenicity in children and macaques demyelinating diseases
Authors
Che Serguera
Lev Stimmer
Claire-Maelle Fovet
Philippe Horellou
Vanessa Contreras
Nicolas Tchitchek
Julie Massonneau
Carole Leroy
Audrey Perrin
Julien Flament
Philippe Hantraye
Joanna Demilly
Romain Marignier
Pascale Chrétien
Bert‘t Hart
Jean Boutonnat
Clovis Adam
Roger Le-Grand
Kumaran Deiva
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2019
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1637-7

Other articles of this Issue 1/2019

Journal of Neuroinflammation 1/2019 Go to the issue