Cargando…

Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor

BACKGROUND: Intravenous immunoglobulin (IVIg) has been used to treat a variety of autoimmune disorders including multiple sclerosis (MS); however its mechanism of action remains elusive. Recent work has shown that interleukin-11 (IL-11) mRNAs are upregulated by IVIg in MS patient T cells. Both IVIg...

Descripción completa

Detalles Bibliográficos
Autores principales: Figueiredo, Carlyn A., Drohomyrecky, Paulina C., McCarthy, Stephen D. S., Leontyev, Danila, Ma, Xue-Zhong, Branch, Donald R., Dunn, Shannon E.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4117465/
https://www.ncbi.nlm.nih.gov/pubmed/25078447
http://dx.doi.org/10.1371/journal.pone.0101947
_version_ 1782328701004808192
author Figueiredo, Carlyn A.
Drohomyrecky, Paulina C.
McCarthy, Stephen D. S.
Leontyev, Danila
Ma, Xue-Zhong
Branch, Donald R.
Dunn, Shannon E.
author_facet Figueiredo, Carlyn A.
Drohomyrecky, Paulina C.
McCarthy, Stephen D. S.
Leontyev, Danila
Ma, Xue-Zhong
Branch, Donald R.
Dunn, Shannon E.
author_sort Figueiredo, Carlyn A.
collection PubMed
description BACKGROUND: Intravenous immunoglobulin (IVIg) has been used to treat a variety of autoimmune disorders including multiple sclerosis (MS); however its mechanism of action remains elusive. Recent work has shown that interleukin-11 (IL-11) mRNAs are upregulated by IVIg in MS patient T cells. Both IVIg and IL-11 have been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The objective of this study was to determine whether the protective effects of IVIg in EAE occur through an IL-11 and IL-11 receptor (IL-11R)-dependent mechanism. METHODS: We measured IL-11 in the circulation of mice and IL-11 mRNA expression in various organs after IVIg treatment. We then followed with EAE studies to test the efficacy of IVIg in wild-type (WT) mice and in mice deficient for the IL-11 receptor (IL-11Rα(−/−)). Furthermore, we evaluated myelin-specific Th1 and Th17 responses and assessed spinal cord inflammation and demyelination in WT and IL-11Rα(−/−) mice, with and without IVIg treatment. We also examined the direct effects of mouse recombinant IL-11 on the production of IL-17 by lymph node mononuclear cells. RESULTS: IVIg treatment induced a dramatic surge (>1000-fold increase) in the levels of IL-11 in the circulation and a prominent increase of IL-11 mRNA expression in the liver. Furthermore, we found that IL-11Rα(−/−) mice, unlike WT mice, although initially protected, were resistant to full protection by IVIg during EAE and developed disease with a similar incidence and severity as control-treated IL-11Rα(−/−) mice, despite initially showing protection. We observed that Th17 cytokine production by myelin-reactive T cells in the draining lymph nodes was unaffected by IVIg in IL-11Rα(−/−) mice, yet was downregulated in WT mice. Finally, IL-11 was shown to directly inhibit IL-17 production of lymph node cells in culture. CONCLUSION: These results implicate IL-11 as an important immune effector of IVIg in the prevention of Th17-mediated autoimmune inflammation during EAE.
format Online
Article
Text
id pubmed-4117465
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-41174652014-08-04 Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor Figueiredo, Carlyn A. Drohomyrecky, Paulina C. McCarthy, Stephen D. S. Leontyev, Danila Ma, Xue-Zhong Branch, Donald R. Dunn, Shannon E. PLoS One Research Article BACKGROUND: Intravenous immunoglobulin (IVIg) has been used to treat a variety of autoimmune disorders including multiple sclerosis (MS); however its mechanism of action remains elusive. Recent work has shown that interleukin-11 (IL-11) mRNAs are upregulated by IVIg in MS patient T cells. Both IVIg and IL-11 have been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The objective of this study was to determine whether the protective effects of IVIg in EAE occur through an IL-11 and IL-11 receptor (IL-11R)-dependent mechanism. METHODS: We measured IL-11 in the circulation of mice and IL-11 mRNA expression in various organs after IVIg treatment. We then followed with EAE studies to test the efficacy of IVIg in wild-type (WT) mice and in mice deficient for the IL-11 receptor (IL-11Rα(−/−)). Furthermore, we evaluated myelin-specific Th1 and Th17 responses and assessed spinal cord inflammation and demyelination in WT and IL-11Rα(−/−) mice, with and without IVIg treatment. We also examined the direct effects of mouse recombinant IL-11 on the production of IL-17 by lymph node mononuclear cells. RESULTS: IVIg treatment induced a dramatic surge (>1000-fold increase) in the levels of IL-11 in the circulation and a prominent increase of IL-11 mRNA expression in the liver. Furthermore, we found that IL-11Rα(−/−) mice, unlike WT mice, although initially protected, were resistant to full protection by IVIg during EAE and developed disease with a similar incidence and severity as control-treated IL-11Rα(−/−) mice, despite initially showing protection. We observed that Th17 cytokine production by myelin-reactive T cells in the draining lymph nodes was unaffected by IVIg in IL-11Rα(−/−) mice, yet was downregulated in WT mice. Finally, IL-11 was shown to directly inhibit IL-17 production of lymph node cells in culture. CONCLUSION: These results implicate IL-11 as an important immune effector of IVIg in the prevention of Th17-mediated autoimmune inflammation during EAE. Public Library of Science 2014-07-31 /pmc/articles/PMC4117465/ /pubmed/25078447 http://dx.doi.org/10.1371/journal.pone.0101947 Text en © 2014 Figueiredo et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Figueiredo, Carlyn A.
Drohomyrecky, Paulina C.
McCarthy, Stephen D. S.
Leontyev, Danila
Ma, Xue-Zhong
Branch, Donald R.
Dunn, Shannon E.
Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title_full Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title_fullStr Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title_full_unstemmed Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title_short Optimal Attenuation of Experimental Autoimmune Encephalomyelitis by Intravenous Immunoglobulin Requires an Intact Interleukin-11 Receptor
title_sort optimal attenuation of experimental autoimmune encephalomyelitis by intravenous immunoglobulin requires an intact interleukin-11 receptor
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4117465/
https://www.ncbi.nlm.nih.gov/pubmed/25078447
http://dx.doi.org/10.1371/journal.pone.0101947
work_keys_str_mv AT figueiredocarlyna optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT drohomyreckypaulinac optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT mccarthystephends optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT leontyevdanila optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT maxuezhong optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT branchdonaldr optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor
AT dunnshannone optimalattenuationofexperimentalautoimmuneencephalomyelitisbyintravenousimmunoglobulinrequiresanintactinterleukin11receptor