Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Protection from experimental autoimmune encephalomyelitis by polyclonal IgG requires adjuvant-induced inflammation

Authors: Isaak Quast, Christian W. Keller, Patrick Weber, Christoph Schneider, Stephan von Gunten, Jan D. Lünemann

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Intravenous immunoglobulin (IVIG) proved to be an efficient anti-inflammatory treatment for a growing number of neuroinflammatory diseases and protects against the development of experimental autoimmune encephalomyelitis (EAE), a widely used animal model for multiple sclerosis (MS).

Methods

The clinical efficacy of IVIG and IVIG-derived F(ab’)2 fragments, generated using the streptococcal cysteine proteinase Ide-S, was evaluated in EAE induced by active immunization and by adoptive transfer of myelin-specific T cells. Frequency, phenotype, and functional characteristics of T cell subsets and myeloid cells were determined by flow cytometry. Antibody binding to microbial antigen and cytokine production by innate immune cells was assessed by ELISA.

Results

We report that the protective effect of IVIG is lost in the adoptive transfer model of EAE and requires prophylactic administration during disease induction. IVIG-derived Fc fragments are not required for protection against EAE, since administration of F(ab’)2 fragments fully recapitulated the clinical efficacy of IVIG. F(ab’)2-treated mice showed a substantial decrease in splenic effector T cell expansion and cytokine production (GM-CSF, IFN-γ, IL-17A) 9 days after immunization. Inhibition of effector T cell responses was not associated with an increase in total numbers of Tregs but with decreased activation of innate myeloid cells such as neutrophils, monocytes, and dendritic cells. Therapeutically effective IVIG-derived F(ab’)2 fragments inhibited adjuvant-induced innate immune cell activation as determined by IL-12/23 p40 production and recognized mycobacterial antigens contained in Freund’s complete adjuvant which is required for induction of active EAE.

Conclusions

Our data indicate that F(ab’)2-mediated neutralization of adjuvant contributes to the therapeutic efficacy of anti-inflammatory IgG. These findings might partly explain the discrepancy of IVIG efficacy in EAE and MS.
Literature
1.
go back to reference Lunemann JD, Nimmerjahn F, Dalakas MC. Intravenous immunoglobulin in neurology—mode of action and clinical efficacy. Nat Rev Neurol. 2015;11:80–9.CrossRefPubMed Lunemann JD, Nimmerjahn F, Dalakas MC. Intravenous immunoglobulin in neurology—mode of action and clinical efficacy. Nat Rev Neurol. 2015;11:80–9.CrossRefPubMed
2.
go back to reference Achiron A, Margalit R, Hershkoviz R, Markovits D, Reshef T, Melamed E, et al. Intravenous immunoglobulin treatment of experimental T cell-mediated autoimmune disease. Upregulation of T cell proliferation and downregulation of tumor necrosis factor alpha secretion. J Clin Invest. 1994;93:600–5.PubMedCentralCrossRefPubMed Achiron A, Margalit R, Hershkoviz R, Markovits D, Reshef T, Melamed E, et al. Intravenous immunoglobulin treatment of experimental T cell-mediated autoimmune disease. Upregulation of T cell proliferation and downregulation of tumor necrosis factor alpha secretion. J Clin Invest. 1994;93:600–5.PubMedCentralCrossRefPubMed
3.
go back to reference Othy S, Topcu S, Saha C, Kothapalli P, Lacroix-Desmazes S, Kasermann F, et al. Sialylation may be dispensable for reciprocal modulation of helper T cells by intravenous immunoglobulin. Eur J Immunol. 2014;44:2059–63.CrossRefPubMed Othy S, Topcu S, Saha C, Kothapalli P, Lacroix-Desmazes S, Kasermann F, et al. Sialylation may be dispensable for reciprocal modulation of helper T cells by intravenous immunoglobulin. Eur J Immunol. 2014;44:2059–63.CrossRefPubMed
4.
go back to reference Trinath J, Hegde P, Sharma M, Maddur MS, Rabin M, Vallat JM, et al. Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells. Blood. 2013;122:1419–27.CrossRefPubMed Trinath J, Hegde P, Sharma M, Maddur MS, Rabin M, Vallat JM, et al. Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells. Blood. 2013;122:1419–27.CrossRefPubMed
5.
go back to reference Humle Jorgensen S, Sorensen PS. Intravenous immunoglobulin treatment of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis. J Neurol Sci. 2005;233:61–5.CrossRefPubMed Humle Jorgensen S, Sorensen PS. Intravenous immunoglobulin treatment of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis. J Neurol Sci. 2005;233:61–5.CrossRefPubMed
6.
go back to reference Jorgensen SH, Storm N, Jensen PE, Laursen H, Sorensen PS. IVIG enters the central nervous system during treatment of experimental autoimmune encephalomyelitis and is localised to inflammatory lesions. Exp Brain Res. 2007;178:462–9.CrossRefPubMed Jorgensen SH, Storm N, Jensen PE, Laursen H, Sorensen PS. IVIG enters the central nervous system during treatment of experimental autoimmune encephalomyelitis and is localised to inflammatory lesions. Exp Brain Res. 2007;178:462–9.CrossRefPubMed
7.
go back to reference Ephrem A, Chamat S, Miquel C, Fisson S, Mouthon L, Caligiuri G, et al. Expansion of CD4 + CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis. Blood. 2008;111:715–22.CrossRefPubMed Ephrem A, Chamat S, Miquel C, Fisson S, Mouthon L, Caligiuri G, et al. Expansion of CD4 + CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis. Blood. 2008;111:715–22.CrossRefPubMed
8.
go back to reference Fiebiger BM, Maamary J, Pincetic A, Ravetch JV. Protection in antibody- and T cell-mediated autoimmune diseases by antiinflammatory IgG Fcs requires type II FcRs. Proc Natl Acad Sci U S A. 2015;112:E2385–94.PubMedCentralCrossRefPubMed Fiebiger BM, Maamary J, Pincetic A, Ravetch JV. Protection in antibody- and T cell-mediated autoimmune diseases by antiinflammatory IgG Fcs requires type II FcRs. Proc Natl Acad Sci U S A. 2015;112:E2385–94.PubMedCentralCrossRefPubMed
9.
go back to reference Sorensen PS, Haas J, Sellebjerg F, Olsson T, Ravnborg M, Group TS. IV immunoglobulins as add-on treatment to methylprednisolone for acute relapses in MS. Neurology. 2004;63:2028–33.CrossRefPubMed Sorensen PS, Haas J, Sellebjerg F, Olsson T, Ravnborg M, Group TS. IV immunoglobulins as add-on treatment to methylprednisolone for acute relapses in MS. Neurology. 2004;63:2028–33.CrossRefPubMed
10.
go back to reference Visser LH, Beekman R, Tijssen CC, Uitdehaag BM, Lee ML, Movig KL, et al. A randomized, double-blind, placebo-controlled pilot study of i.v. immune globulins in combination with i.v. methylprednisolone in the treatment of relapses in patients with MS. Mult Scler. 2004;10:89–91.CrossRefPubMed Visser LH, Beekman R, Tijssen CC, Uitdehaag BM, Lee ML, Movig KL, et al. A randomized, double-blind, placebo-controlled pilot study of i.v. immune globulins in combination with i.v. methylprednisolone in the treatment of relapses in patients with MS. Mult Scler. 2004;10:89–91.CrossRefPubMed
11.
go back to reference Hommes OR, Sorensen PS, Fazekas F, Enriquez MM, Koelmel HW, Fernandez O, et al. Intravenous immunoglobulin in secondary progressive multiple sclerosis: randomised placebo-controlled trial. Lancet. 2004;364:1149–56.CrossRefPubMed Hommes OR, Sorensen PS, Fazekas F, Enriquez MM, Koelmel HW, Fernandez O, et al. Intravenous immunoglobulin in secondary progressive multiple sclerosis: randomised placebo-controlled trial. Lancet. 2004;364:1149–56.CrossRefPubMed
12.
go back to reference Fazekas F, Lublin FD, Li D, Freedman MS, Hartung HP, Rieckmann P, et al. Intravenous immunoglobulin in relapsing-remitting multiple sclerosis: a dose-finding trial. Neurology. 2008;71:265–71.CrossRefPubMed Fazekas F, Lublin FD, Li D, Freedman MS, Hartung HP, Rieckmann P, et al. Intravenous immunoglobulin in relapsing-remitting multiple sclerosis: a dose-finding trial. Neurology. 2008;71:265–71.CrossRefPubMed
13.
go back to reference Lünemann JD, Quast I, Dalakas MC. Efficacy of Intravenous Immunoglobulin in Neurological Diseases. Neurotherapeutics. 2016;13:34–46.CrossRefPubMed Lünemann JD, Quast I, Dalakas MC. Efficacy of Intravenous Immunoglobulin in Neurological Diseases. Neurotherapeutics. 2016;13:34–46.CrossRefPubMed
14.
go back to reference Gold R, Stangel M, Dalakas MC. Drug Insight: the use of intravenous immunoglobulin in neurology—therapeutic considerations and practical issues. Nat Clin Pract Neurol. 2007;3:36–44.CrossRefPubMed Gold R, Stangel M, Dalakas MC. Drug Insight: the use of intravenous immunoglobulin in neurology—therapeutic considerations and practical issues. Nat Clin Pract Neurol. 2007;3:36–44.CrossRefPubMed
15.
go back to reference Anthony RM, Wermeling F, Karlsson MC, Ravetch JV. Identification of a receptor required for the anti-inflammatory activity of IVIG. Proc Natl Acad Sci U S A. 2008;105:19571–8.PubMedCentralCrossRefPubMed Anthony RM, Wermeling F, Karlsson MC, Ravetch JV. Identification of a receptor required for the anti-inflammatory activity of IVIG. Proc Natl Acad Sci U S A. 2008;105:19571–8.PubMedCentralCrossRefPubMed
16.
go back to reference Bruhns P, Samuelsson A, Pollard JW, Ravetch JV. Colony-stimulating factor-1-dependent macrophages are responsible for IVIG protection in antibody-induced autoimmune disease. Immunity. 2003;18:573–81.CrossRefPubMed Bruhns P, Samuelsson A, Pollard JW, Ravetch JV. Colony-stimulating factor-1-dependent macrophages are responsible for IVIG protection in antibody-induced autoimmune disease. Immunity. 2003;18:573–81.CrossRefPubMed
17.
go back to reference Kaneko Y, Nimmerjahn F, Madaio MP, Ravetch JV. Pathology and protection in nephrotoxic nephritis is determined by selective engagement of specific Fc receptors. J Exp Med. 2006;203:789–97.PubMedCentralCrossRefPubMed Kaneko Y, Nimmerjahn F, Madaio MP, Ravetch JV. Pathology and protection in nephrotoxic nephritis is determined by selective engagement of specific Fc receptors. J Exp Med. 2006;203:789–97.PubMedCentralCrossRefPubMed
18.
go back to reference Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science. 2006;313:670–3.CrossRefPubMed Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science. 2006;313:670–3.CrossRefPubMed
19.
go back to reference Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science. 2001;291:484–6.CrossRefPubMed Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science. 2001;291:484–6.CrossRefPubMed
20.
go back to reference von Pawel-Rammingen U, Johansson BP, Bjorck L. IdeS, a novel streptococcal cysteine proteinase with unique specificity for immunoglobulin G. EMBO J. 2002;21:1607–15.CrossRef von Pawel-Rammingen U, Johansson BP, Bjorck L. IdeS, a novel streptococcal cysteine proteinase with unique specificity for immunoglobulin G. EMBO J. 2002;21:1607–15.CrossRef
21.
go back to reference Bettelli E, Pagany M, Weiner HL, Linington C, Sobel RA, Kuchroo VK. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J Exp Med. 2003;197:1073–81.PubMedCentralCrossRefPubMed Bettelli E, Pagany M, Weiner HL, Linington C, Sobel RA, Kuchroo VK. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J Exp Med. 2003;197:1073–81.PubMedCentralCrossRefPubMed
22.
go back to reference Miller SD, Karpus WJ. Experimental autoimmune encephalomyelitis in the mouse. Curr Protoc Immunol. 2007;Chapter 15:Unit 15 11. Miller SD, Karpus WJ. Experimental autoimmune encephalomyelitis in the mouse. Curr Protoc Immunol. 2007;Chapter 15:Unit 15 11.
23.
go back to reference Frommer F, Heinen TJ, Wunderlich FT, Yogev N, Buch T, Roers A, et al. Tolerance without clonal expansion: self-antigen-expressing B cells program self-reactive T cells for future deletion. J Immunol. 2008;181:5748–59.CrossRefPubMed Frommer F, Heinen TJ, Wunderlich FT, Yogev N, Buch T, Roers A, et al. Tolerance without clonal expansion: self-antigen-expressing B cells program self-reactive T cells for future deletion. J Immunol. 2008;181:5748–59.CrossRefPubMed
24.
go back to reference Stromnes IM, Goverman JM. Passive induction of experimental allergic encephalomyelitis. Nat Protoc. 2006;1:1952–60.CrossRefPubMed Stromnes IM, Goverman JM. Passive induction of experimental allergic encephalomyelitis. Nat Protoc. 2006;1:1952–60.CrossRefPubMed
25.
go back to reference Codarri L, Gyulveszi G, Tosevski V, Hesske L, Fontana A, Magnenat L, et al. RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011;12:560–7.CrossRefPubMed Codarri L, Gyulveszi G, Tosevski V, Hesske L, Fontana A, Magnenat L, et al. RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011;12:560–7.CrossRefPubMed
26.
go back to reference Geering B, Gurzeler U, Federzoni E, Kaufmann T, Simon HU. A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils. Blood. 2011;117:5953–62.CrossRefPubMed Geering B, Gurzeler U, Federzoni E, Kaufmann T, Simon HU. A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils. Blood. 2011;117:5953–62.CrossRefPubMed
27.
go back to reference Wehrli M, Cortinas-Elizondo F, Hlushchuk R, Daudel F, Villiger PM, Miescher S, et al. Human IgA Fc receptor FcalphaRI (CD89) triggers different forms of neutrophil death depending on the inflammatory microenvironment. J Immunol. 2014;193:5649–59.CrossRefPubMed Wehrli M, Cortinas-Elizondo F, Hlushchuk R, Daudel F, Villiger PM, Miescher S, et al. Human IgA Fc receptor FcalphaRI (CD89) triggers different forms of neutrophil death depending on the inflammatory microenvironment. J Immunol. 2014;193:5649–59.CrossRefPubMed
28.
go back to reference von Gunten S, Schaub A, Vogel M, Stadler BM, Miescher S, Simon HU. Immunologic and functional evidence for anti-Siglec-9 autoantibodies in intravenous immunoglobulin preparations. Blood. 2006;108:4255–9.CrossRef von Gunten S, Schaub A, Vogel M, Stadler BM, Miescher S, Simon HU. Immunologic and functional evidence for anti-Siglec-9 autoantibodies in intravenous immunoglobulin preparations. Blood. 2006;108:4255–9.CrossRef
29.
go back to reference Croxford AL, Lanzinger M, Hartmann FJ, Schreiner B, Mair F, Pelczar P, et al. The cytokine GM-CSF drives the inflammatory signature of CCR2(+) monocytes and licenses autoimmunity. Immunity. 2015;43:502–14.CrossRefPubMed Croxford AL, Lanzinger M, Hartmann FJ, Schreiner B, Mair F, Pelczar P, et al. The cytokine GM-CSF drives the inflammatory signature of CCR2(+) monocytes and licenses autoimmunity. Immunity. 2015;43:502–14.CrossRefPubMed
30.
go back to reference Billiau A, Matthys P. Modes of action of Freund’s adjuvants in experimental models of autoimmune diseases. J Leukoc Biol. 2001;70:849–60.PubMed Billiau A, Matthys P. Modes of action of Freund’s adjuvants in experimental models of autoimmune diseases. J Leukoc Biol. 2001;70:849–60.PubMed
31.
go back to reference Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–40.PubMedCentralCrossRefPubMed Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–40.PubMedCentralCrossRefPubMed
32.
go back to reference Lassmann H, Zimprich F, Rossler K, Vass K. Inflammation in the nervous system. Basic mechanisms and immunological concepts. Rev Neurol (Paris). 1991;147:763–81. Lassmann H, Zimprich F, Rossler K, Vass K. Inflammation in the nervous system. Basic mechanisms and immunological concepts. Rev Neurol (Paris). 1991;147:763–81.
33.
go back to reference Steinbach K, Piedavent M, Bauer S, Neumann JT, Friese MA. Neutrophils amplify autoimmune central nervous system infiltrates by maturing local APCs. J Immunol. 2013;191:4531–9.CrossRefPubMed Steinbach K, Piedavent M, Bauer S, Neumann JT, Friese MA. Neutrophils amplify autoimmune central nervous system infiltrates by maturing local APCs. J Immunol. 2013;191:4531–9.CrossRefPubMed
34.
go back to reference Altznauer F, von Gunten S, Spath P, Simon HU. Concurrent presence of agonistic and antagonistic anti-CD95 autoantibodies in intravenous Ig preparations. J Allergy Clin Immunol. 2003;112:1185–90.CrossRefPubMed Altznauer F, von Gunten S, Spath P, Simon HU. Concurrent presence of agonistic and antagonistic anti-CD95 autoantibodies in intravenous Ig preparations. J Allergy Clin Immunol. 2003;112:1185–90.CrossRefPubMed
35.
go back to reference von Gunten S, Yousefi S, Seitz M, Jakob SM, Schaffner T, Seger R, et al. Siglec-9 transduces apoptotic and nonapoptotic death signals into neutrophils depending on the proinflammatory cytokine environment. Blood. 2005;106:1423–31.CrossRef von Gunten S, Yousefi S, Seitz M, Jakob SM, Schaffner T, Seger R, et al. Siglec-9 transduces apoptotic and nonapoptotic death signals into neutrophils depending on the proinflammatory cytokine environment. Blood. 2005;106:1423–31.CrossRef
36.
go back to reference Aoyama-Ishikawa M, Seishu A, Kawakami S, Maeshige N, Miyoshi M, Ueda T, et al. Intravenous immunoglobulin-induced neutrophil apoptosis in the lung during murine endotoxemia. Surg Infect (Larchmt). 2014;15:36–42.CrossRef Aoyama-Ishikawa M, Seishu A, Kawakami S, Maeshige N, Miyoshi M, Ueda T, et al. Intravenous immunoglobulin-induced neutrophil apoptosis in the lung during murine endotoxemia. Surg Infect (Larchmt). 2014;15:36–42.CrossRef
37.
go back to reference Lamari F, Karamanos NK, Papadopoulou-Alataki E, Kanakoudi-Tsakalidou F, Dimitracopoulos G, Anastassiou ED. Monitoring of two intravenous immunoglobulin. Preparations for immunoglobulin G subclasses and specific antibodies to bacterial surface antigens and relation with their levels in treated immunodeficient patients. J Pharm Biomed Anal. 2000;22:1029–36.CrossRefPubMed Lamari F, Karamanos NK, Papadopoulou-Alataki E, Kanakoudi-Tsakalidou F, Dimitracopoulos G, Anastassiou ED. Monitoring of two intravenous immunoglobulin. Preparations for immunoglobulin G subclasses and specific antibodies to bacterial surface antigens and relation with their levels in treated immunodeficient patients. J Pharm Biomed Anal. 2000;22:1029–36.CrossRefPubMed
38.
go back to reference Schneider C, Smith DF, Cummings RD, Boligan KF, Hamilton RG, Bochner BS, et al. The human IgG anti-carbohydrate repertoire exhibits a universal architecture and contains specificity for microbial attachment sites. Sci Transl Med. 2015;7:269ra261.CrossRef Schneider C, Smith DF, Cummings RD, Boligan KF, Hamilton RG, Bochner BS, et al. The human IgG anti-carbohydrate repertoire exhibits a universal architecture and contains specificity for microbial attachment sites. Sci Transl Med. 2015;7:269ra261.CrossRef
39.
go back to reference Roy E, Stavropoulos E, Brennan J, Coade S, Grigorieva E, Walker B, et al. Therapeutic efficacy of high-dose intravenous immunoglobulin in Mycobacterium tuberculosis infection in mice. Infect Immun. 2005;73:6101–9.PubMedCentralCrossRefPubMed Roy E, Stavropoulos E, Brennan J, Coade S, Grigorieva E, Walker B, et al. Therapeutic efficacy of high-dose intravenous immunoglobulin in Mycobacterium tuberculosis infection in mice. Infect Immun. 2005;73:6101–9.PubMedCentralCrossRefPubMed
40.
go back to reference Thakker P, Leach MW, Kuang W, Benoit SE, Leonard JP, Marusic S. IL-23 is critical in the induction but not in the effector phase of experimental autoimmune encephalomyelitis. J Immunol. 2007;178:2589–98.CrossRefPubMed Thakker P, Leach MW, Kuang W, Benoit SE, Leonard JP, Marusic S. IL-23 is critical in the induction but not in the effector phase of experimental autoimmune encephalomyelitis. J Immunol. 2007;178:2589–98.CrossRefPubMed
41.
go back to reference Jorgensen SH, Jensen PE, Laursen H, Sorensen PS. Intravenous immunoglobulin ameliorates experimental autoimmune encephalomyelitis and reduces neuropathological abnormalities when administered prophylactically. Neurol Res. 2005;27:591–7.CrossRefPubMed Jorgensen SH, Jensen PE, Laursen H, Sorensen PS. Intravenous immunoglobulin ameliorates experimental autoimmune encephalomyelitis and reduces neuropathological abnormalities when administered prophylactically. Neurol Res. 2005;27:591–7.CrossRefPubMed
42.
go back to reference Schluesener HJ, Sobel RA, Linington C, Weiner HL. A monoclonal antibody against a myelin oligodendrocyte glycoprotein induces relapses and demyelination in central nervous system autoimmune disease. J Immunol. 1987;139:4016–21.PubMed Schluesener HJ, Sobel RA, Linington C, Weiner HL. A monoclonal antibody against a myelin oligodendrocyte glycoprotein induces relapses and demyelination in central nervous system autoimmune disease. J Immunol. 1987;139:4016–21.PubMed
43.
44.
go back to reference Castro LH, Ropper AH. Human immune globulin infusion in Guillain-Barre syndrome: worsening during and after treatment. Neurology. 1993;43:1034–6.CrossRefPubMed Castro LH, Ropper AH. Human immune globulin infusion in Guillain-Barre syndrome: worsening during and after treatment. Neurology. 1993;43:1034–6.CrossRefPubMed
45.
go back to reference Irani DN, Cornblath DR, Chaudhry V, Borel C, Hanley DF. Relapse in Guillain-Barre syndrome after treatment with human immune globulin. Neurology. 1993;43:872–5.CrossRefPubMed Irani DN, Cornblath DR, Chaudhry V, Borel C, Hanley DF. Relapse in Guillain-Barre syndrome after treatment with human immune globulin. Neurology. 1993;43:872–5.CrossRefPubMed
46.
go back to reference Gelfand EW. Differences between IGIV products: impact on clinical outcome. Int Immunopharmacol. 2006;6:592–9.CrossRefPubMed Gelfand EW. Differences between IGIV products: impact on clinical outcome. Int Immunopharmacol. 2006;6:592–9.CrossRefPubMed
47.
go back to reference Achiron A, Gilad R, Margalit R, Gabbay U, Sarova-Pinhas I, Cohen IR, et al. Intravenous gammaglobulin treatment in multiple sclerosis and experimental autoimmune encephalomyelitis: delineation of usage and mode of action. J Neurol Neurosurg Psychiatry. 1994;57(Suppl):57–61.PubMedCentralCrossRefPubMed Achiron A, Gilad R, Margalit R, Gabbay U, Sarova-Pinhas I, Cohen IR, et al. Intravenous gammaglobulin treatment in multiple sclerosis and experimental autoimmune encephalomyelitis: delineation of usage and mode of action. J Neurol Neurosurg Psychiatry. 1994;57(Suppl):57–61.PubMedCentralCrossRefPubMed
48.
go back to reference Aktas O, Waiczies S, Grieger U, Wendling U, Zschenderlein R, Zipp F. Polyspecific immunoglobulins (IVIg) suppress proliferation of human (auto)antigen-specific T cells without inducing apoptosis. J Neuroimmunol. 2001;114:160–7.CrossRefPubMed Aktas O, Waiczies S, Grieger U, Wendling U, Zschenderlein R, Zipp F. Polyspecific immunoglobulins (IVIg) suppress proliferation of human (auto)antigen-specific T cells without inducing apoptosis. J Neuroimmunol. 2001;114:160–7.CrossRefPubMed
49.
go back to reference Prasad NK, Papoff G, Zeuner A, Bonnin E, Kazatchkine MD, Ruberti G, et al. Therapeutic preparations of normal polyspecific IgG (IVIg) induce apoptosis in human lymphocytes and monocytes: a novel mechanism of action of IVIg involving the Fas apoptotic pathway. J Immunol. 1998;161:3781–90.PubMed Prasad NK, Papoff G, Zeuner A, Bonnin E, Kazatchkine MD, Ruberti G, et al. Therapeutic preparations of normal polyspecific IgG (IVIg) induce apoptosis in human lymphocytes and monocytes: a novel mechanism of action of IVIg involving the Fas apoptotic pathway. J Immunol. 1998;161:3781–90.PubMed
50.
go back to reference Pashov A, Dubey C, Kaveri SV, Lectard B, Huang YM, Kazatchkine MD, et al. Normal immunoglobulin G protects against experimental allergic encephalomyelitis by inducing transferable T cell unresponsiveness to myelin basic protein. Eur J Immunol. 1998;28:1823–31.CrossRefPubMed Pashov A, Dubey C, Kaveri SV, Lectard B, Huang YM, Kazatchkine MD, et al. Normal immunoglobulin G protects against experimental allergic encephalomyelitis by inducing transferable T cell unresponsiveness to myelin basic protein. Eur J Immunol. 1998;28:1823–31.CrossRefPubMed
51.
go back to reference Weishaupt A, Kuhlmann T, Schonrock LM, Toyka KV, Bruck W, Gold R. Effects of intravenous immunoglobulins on T cell and oligodendrocyte apoptosis in high-dose antigen therapy in experimental autoimmune encephalomyelitis. Acta Neuropathol. 2002;104:385–90.PubMed Weishaupt A, Kuhlmann T, Schonrock LM, Toyka KV, Bruck W, Gold R. Effects of intravenous immunoglobulins on T cell and oligodendrocyte apoptosis in high-dose antigen therapy in experimental autoimmune encephalomyelitis. Acta Neuropathol. 2002;104:385–90.PubMed
52.
go back to reference Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol. 2013;13:176–89.CrossRefPubMed Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol. 2013;13:176–89.CrossRefPubMed
53.
go back to reference Anthony RM, Nimmerjahn F, Ashline DJ, Reinhold VN, Paulson JC, Ravetch JV. Recapitulation of IVIG anti-inflammatory activity with a recombinant IgG Fc. Science. 2008;320:373–6.PubMedCentralCrossRefPubMed Anthony RM, Nimmerjahn F, Ashline DJ, Reinhold VN, Paulson JC, Ravetch JV. Recapitulation of IVIG anti-inflammatory activity with a recombinant IgG Fc. Science. 2008;320:373–6.PubMedCentralCrossRefPubMed
54.
go back to reference Washburn N, Schwab I, Ortiz D, Bhatnagar N, Lansing JC, Medeiros A, et al. Controlled tetra-Fc sialylation of IVIg results in a drug candidate with consistent enhanced anti-inflammatory activity. Proc Natl Acad Sci U S A. 2015;112:E1297–306.PubMedCentralCrossRefPubMed Washburn N, Schwab I, Ortiz D, Bhatnagar N, Lansing JC, Medeiros A, et al. Controlled tetra-Fc sialylation of IVIg results in a drug candidate with consistent enhanced anti-inflammatory activity. Proc Natl Acad Sci U S A. 2015;112:E1297–306.PubMedCentralCrossRefPubMed
55.
go back to reference Baudino L, Nimmerjahn F, Azeredo da Silveira S, Martinez-Soria E, Saito T, Carroll M, et al. Differential contribution of three activating IgG Fc receptors (FcgammaRI, FcgammaRIII, and FcgammaRIV) to IgG2a- and IgG2b-induced autoimmune hemolytic anemia in mice. J Immunol. 2008;180:1948–53.CrossRefPubMed Baudino L, Nimmerjahn F, Azeredo da Silveira S, Martinez-Soria E, Saito T, Carroll M, et al. Differential contribution of three activating IgG Fc receptors (FcgammaRI, FcgammaRIII, and FcgammaRIV) to IgG2a- and IgG2b-induced autoimmune hemolytic anemia in mice. J Immunol. 2008;180:1948–53.CrossRefPubMed
56.
go back to reference Beers SA, French RR, Chan HT, Lim SH, Jarrett TC, Vidal RM, et al. Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection. Blood. 2010;115:5191–201.CrossRefPubMed Beers SA, French RR, Chan HT, Lim SH, Jarrett TC, Vidal RM, et al. Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection. Blood. 2010;115:5191–201.CrossRefPubMed
57.
go back to reference Biburger M, Aschermann S, Schwab I, Lux A, Albert H, Danzer H, et al. Monocyte subsets responsible for immunoglobulin G-dependent effector functions in vivo. Immunity. 2011;35:932–44.CrossRefPubMed Biburger M, Aschermann S, Schwab I, Lux A, Albert H, Danzer H, et al. Monocyte subsets responsible for immunoglobulin G-dependent effector functions in vivo. Immunity. 2011;35:932–44.CrossRefPubMed
58.
go back to reference Clynes R, Ravetch JV. Cytotoxic antibodies trigger inflammation through Fc receptors. Immunity. 1995;3:21–6.CrossRefPubMed Clynes R, Ravetch JV. Cytotoxic antibodies trigger inflammation through Fc receptors. Immunity. 1995;3:21–6.CrossRefPubMed
59.
go back to reference Fossati-Jimack L, Ioan-Facsinay A, Reininger L, Chicheportiche Y, Watanabe N, Saito T, et al. Markedly different pathogenicity of four immunoglobulin G isotype-switch variants of an antierythrocyte autoantibody is based on their capacity to interact in vivo with the low-affinity Fcgamma receptor III. J Exp Med. 2000;191:1293–302.PubMedCentralCrossRefPubMed Fossati-Jimack L, Ioan-Facsinay A, Reininger L, Chicheportiche Y, Watanabe N, Saito T, et al. Markedly different pathogenicity of four immunoglobulin G isotype-switch variants of an antierythrocyte autoantibody is based on their capacity to interact in vivo with the low-affinity Fcgamma receptor III. J Exp Med. 2000;191:1293–302.PubMedCentralCrossRefPubMed
60.
go back to reference Hamaguchi Y, Xiu Y, Komura K, Nimmerjahn F, Tedder TF. Antibody isotype-specific engagement of Fcgamma receptors regulates B lymphocyte depletion during CD20 immunotherapy. J Exp Med. 2006;203:743–53.PubMedCentralCrossRefPubMed Hamaguchi Y, Xiu Y, Komura K, Nimmerjahn F, Tedder TF. Antibody isotype-specific engagement of Fcgamma receptors regulates B lymphocyte depletion during CD20 immunotherapy. J Exp Med. 2006;203:743–53.PubMedCentralCrossRefPubMed
61.
go back to reference Hogarth PM. Fc receptors are major mediators of antibody based inflammation in autoimmunity. Curr Opin Immunol. 2002;14:798–802.CrossRefPubMed Hogarth PM. Fc receptors are major mediators of antibody based inflammation in autoimmunity. Curr Opin Immunol. 2002;14:798–802.CrossRefPubMed
62.
go back to reference Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity. 2005;23:41–51.CrossRefPubMed Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity. 2005;23:41–51.CrossRefPubMed
63.
go back to reference Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 2005;310:1510–2.CrossRefPubMed Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 2005;310:1510–2.CrossRefPubMed
64.
go back to reference Ravetch JV, Clynes RA. Divergent roles for Fc receptors and complement in vivo. Annu Rev Immunol. 1998;16:421–32.CrossRefPubMed Ravetch JV, Clynes RA. Divergent roles for Fc receptors and complement in vivo. Annu Rev Immunol. 1998;16:421–32.CrossRefPubMed
65.
go back to reference Sylvestre D, Clynes R, Ma M, Warren H, Carroll MC, Ravetch JV. Immunoglobulin G-mediated inflammatory responses develop normally in complement-deficient mice. J Exp Med. 1996;184:2385–92.PubMedCentralCrossRefPubMed Sylvestre D, Clynes R, Ma M, Warren H, Carroll MC, Ravetch JV. Immunoglobulin G-mediated inflammatory responses develop normally in complement-deficient mice. J Exp Med. 1996;184:2385–92.PubMedCentralCrossRefPubMed
66.
go back to reference Sylvestre DL, Ravetch JV. Fc receptors initiate the Arthus reaction: redefining the inflammatory cascade. Science. 1994;265:1095–8.CrossRefPubMed Sylvestre DL, Ravetch JV. Fc receptors initiate the Arthus reaction: redefining the inflammatory cascade. Science. 1994;265:1095–8.CrossRefPubMed
67.
go back to reference Takai T, Li M, Sylvestre D, Clynes R, Ravetch JV. FcR gamma chain deletion results in pleiotrophic effector cell defects. Cell. 1994;76:519–29.CrossRefPubMed Takai T, Li M, Sylvestre D, Clynes R, Ravetch JV. FcR gamma chain deletion results in pleiotrophic effector cell defects. Cell. 1994;76:519–29.CrossRefPubMed
68.
go back to reference Uchida J, Hamaguchi Y, Oliver JA, Ravetch JV, Poe JC, Haas KM, et al. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J Exp Med. 2004;199:1659–69.PubMedCentralCrossRefPubMed Uchida J, Hamaguchi Y, Oliver JA, Ravetch JV, Poe JC, Haas KM, et al. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J Exp Med. 2004;199:1659–69.PubMedCentralCrossRefPubMed
69.
go back to reference Hjelmstrom P, Juedes AE, Fjell J, Ruddle NH. B-cell-deficient mice develop experimental allergic encephalomyelitis with demyelination after myelin oligodendrocyte glycoprotein sensitization. J Immunol. 1998;161:4480–3.PubMed Hjelmstrom P, Juedes AE, Fjell J, Ruddle NH. B-cell-deficient mice develop experimental allergic encephalomyelitis with demyelination after myelin oligodendrocyte glycoprotein sensitization. J Immunol. 1998;161:4480–3.PubMed
70.
go back to reference Wolf SD, Dittel BN, Hardardottir F, Janeway Jr CA. Experimental autoimmune encephalomyelitis induction in genetically B cell-deficient mice. J Exp Med. 1996;184:2271–8.PubMedCentralCrossRefPubMed Wolf SD, Dittel BN, Hardardottir F, Janeway Jr CA. Experimental autoimmune encephalomyelitis induction in genetically B cell-deficient mice. J Exp Med. 1996;184:2271–8.PubMedCentralCrossRefPubMed
71.
go back to reference Eugster HP, Frei K, Kopf M, Lassmann H, Fontana A. IL-6-deficient mice resist myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. Eur J Immunol. 1998;28:2178–87.CrossRefPubMed Eugster HP, Frei K, Kopf M, Lassmann H, Fontana A. IL-6-deficient mice resist myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. Eur J Immunol. 1998;28:2178–87.CrossRefPubMed
72.
go back to reference Pedotti R, DeVoss JJ, Youssef S, Mitchell D, Wedemeyer J, Madanat R, et al. Multiple elements of the allergic arm of the immune response modulate autoimmune demyelination. Proc Natl Acad Sci U S A. 2003;100:1867–72.PubMedCentralCrossRefPubMed Pedotti R, DeVoss JJ, Youssef S, Mitchell D, Wedemeyer J, Madanat R, et al. Multiple elements of the allergic arm of the immune response modulate autoimmune demyelination. Proc Natl Acad Sci U S A. 2003;100:1867–72.PubMedCentralCrossRefPubMed
73.
go back to reference Urich E, Gutcher I, Prinz M, Becher B. Autoantibody-mediated demyelination depends on complement activation but not activatory Fc-receptors. Proc Natl Acad Sci U S A. 2006;103:18697–702.PubMedCentralCrossRefPubMed Urich E, Gutcher I, Prinz M, Becher B. Autoantibody-mediated demyelination depends on complement activation but not activatory Fc-receptors. Proc Natl Acad Sci U S A. 2006;103:18697–702.PubMedCentralCrossRefPubMed
74.
go back to reference Viard I, Wehrli P, Bullani R, Schneider P, Holler N, Salomon D, et al. Inhibition of toxic epidermal necrolysis by blockade of CD95 with human intravenous immunoglobulin. Science. 1998;282:490–3.CrossRefPubMed Viard I, Wehrli P, Bullani R, Schneider P, Holler N, Salomon D, et al. Inhibition of toxic epidermal necrolysis by blockade of CD95 with human intravenous immunoglobulin. Science. 1998;282:490–3.CrossRefPubMed
75.
go back to reference Bruss JB, Malley R, Halperin S, Dobson S, Dhalla M, McIver J, et al. Treatment of severe pertussis: a study of the safety and pharmacology of intravenous pertussis immunoglobulin. Pediatr Infect Dis J. 1999;18:505–11.CrossRefPubMed Bruss JB, Malley R, Halperin S, Dobson S, Dhalla M, McIver J, et al. Treatment of severe pertussis: a study of the safety and pharmacology of intravenous pertussis immunoglobulin. Pediatr Infect Dis J. 1999;18:505–11.CrossRefPubMed
76.
go back to reference Wu CY, Wang HC, Wang KT, Yang-Chih Shih D, Lo CF, Wang DY. Analyzing titers of antibodies against bacterial and viral antigens, and bacterial toxoids in the intravenous immunoglobulins utilized in Taiwan. Biologicals. 2013;41:88–92.CrossRefPubMed Wu CY, Wang HC, Wang KT, Yang-Chih Shih D, Lo CF, Wang DY. Analyzing titers of antibodies against bacterial and viral antigens, and bacterial toxoids in the intravenous immunoglobulins utilized in Taiwan. Biologicals. 2013;41:88–92.CrossRefPubMed
Metadata
Title
Protection from experimental autoimmune encephalomyelitis by polyclonal IgG requires adjuvant-induced inflammation
Authors
Isaak Quast
Christian W. Keller
Patrick Weber
Christoph Schneider
Stephan von Gunten
Jan D. Lünemann
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0506-x

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue