Cargando…

IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation

See Pluchino and Peruzzotti-Jametti (doi:10.1093/aww266) for a scientific commentary on this article. Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in th...

Descripción completa

Detalles Bibliográficos
Autores principales: Psachoulia, Konstantina, Chamberlain, Kelly A., Heo, Dongeun, Davis, Stephanie E., Paskus, Jeremiah D., Nanescu, Sonia E., Dupree, Jeffrey L., Wynn, Thomas A., Huang, Jeffrey K.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5382940/
https://www.ncbi.nlm.nih.gov/pubmed/27797811
http://dx.doi.org/10.1093/brain/aww254
_version_ 1782520194503016448
author Psachoulia, Konstantina
Chamberlain, Kelly A.
Heo, Dongeun
Davis, Stephanie E.
Paskus, Jeremiah D.
Nanescu, Sonia E.
Dupree, Jeffrey L.
Wynn, Thomas A.
Huang, Jeffrey K.
author_facet Psachoulia, Konstantina
Chamberlain, Kelly A.
Heo, Dongeun
Davis, Stephanie E.
Paskus, Jeremiah D.
Nanescu, Sonia E.
Dupree, Jeffrey L.
Wynn, Thomas A.
Huang, Jeffrey K.
author_sort Psachoulia, Konstantina
collection PubMed
description See Pluchino and Peruzzotti-Jametti (doi:10.1093/aww266) for a scientific commentary on this article. Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in the progressive phase of multiple sclerosis. Emerging evidence indicates that alternatively activated macrophages in central nervous system lesions are required for oligodendrocyte progenitor differentiation into remyelinating oligodendrocytes. Here, we show that an alternatively activated macrophage secreted enzyme, interleukin-four induced one (IL4I1), is upregulated at the onset of inflammation resolution and remyelination in mouse central nervous system lesions after lysolecithin-induced focal demyelination. Focal demyelination in mice lacking IL4I1 or interleukin 4 receptor alpha (IL4Rα) results in increased proinflammatory macrophage density, remyelination impairment, and axonal injury in central nervous system lesions. Conversely, recombinant IL4I1 administration into central nervous system lesions reduces proinflammatory macrophage density, enhances remyelination, and rescues remyelination impairment in IL4Rα deficient mice. We find that IL4I1 does not directly affect oligodendrocyte differentiation, but modulates inflammation by reducing interferon gamma and IL17 expression in lesioned central nervous system tissues, and in activated T cells from splenocyte cultures. Remarkably, intravenous injection of IL4I1 into mice with experimental autoimmune encephalomyelitis at disease onset significantly reversed disease severity, resulting in recovery from hindlimb paralysis. Analysis of post-mortem tissues reveals reduced axonal dystrophy in spinal cord, and decreased CD4(+) T cell populations in spinal cord and spleen tissues. These results indicate that IL4I1 modulates inflammation by regulating T cell expansion, thereby permitting the formation of a favourable environment in the central nervous system tissue for remyelination. Therefore, IL4I1 is a potentially novel therapeutic for promoting central nervous system repair in multiple sclerosis.
format Online
Article
Text
id pubmed-5382940
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-53829402017-04-11 IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation Psachoulia, Konstantina Chamberlain, Kelly A. Heo, Dongeun Davis, Stephanie E. Paskus, Jeremiah D. Nanescu, Sonia E. Dupree, Jeffrey L. Wynn, Thomas A. Huang, Jeffrey K. Brain Original Articles See Pluchino and Peruzzotti-Jametti (doi:10.1093/aww266) for a scientific commentary on this article. Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in the progressive phase of multiple sclerosis. Emerging evidence indicates that alternatively activated macrophages in central nervous system lesions are required for oligodendrocyte progenitor differentiation into remyelinating oligodendrocytes. Here, we show that an alternatively activated macrophage secreted enzyme, interleukin-four induced one (IL4I1), is upregulated at the onset of inflammation resolution and remyelination in mouse central nervous system lesions after lysolecithin-induced focal demyelination. Focal demyelination in mice lacking IL4I1 or interleukin 4 receptor alpha (IL4Rα) results in increased proinflammatory macrophage density, remyelination impairment, and axonal injury in central nervous system lesions. Conversely, recombinant IL4I1 administration into central nervous system lesions reduces proinflammatory macrophage density, enhances remyelination, and rescues remyelination impairment in IL4Rα deficient mice. We find that IL4I1 does not directly affect oligodendrocyte differentiation, but modulates inflammation by reducing interferon gamma and IL17 expression in lesioned central nervous system tissues, and in activated T cells from splenocyte cultures. Remarkably, intravenous injection of IL4I1 into mice with experimental autoimmune encephalomyelitis at disease onset significantly reversed disease severity, resulting in recovery from hindlimb paralysis. Analysis of post-mortem tissues reveals reduced axonal dystrophy in spinal cord, and decreased CD4(+) T cell populations in spinal cord and spleen tissues. These results indicate that IL4I1 modulates inflammation by regulating T cell expansion, thereby permitting the formation of a favourable environment in the central nervous system tissue for remyelination. Therefore, IL4I1 is a potentially novel therapeutic for promoting central nervous system repair in multiple sclerosis. Oxford University Press 2016-12 2016-10-25 /pmc/articles/PMC5382940/ /pubmed/27797811 http://dx.doi.org/10.1093/brain/aww254 Text en © The Author (2016). Published by Oxford University Press on behalf of the Guarantors of Brain. http://creativecommons.org/licenses/by-nc/4.0/ This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
spellingShingle Original Articles
Psachoulia, Konstantina
Chamberlain, Kelly A.
Heo, Dongeun
Davis, Stephanie E.
Paskus, Jeremiah D.
Nanescu, Sonia E.
Dupree, Jeffrey L.
Wynn, Thomas A.
Huang, Jeffrey K.
IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title_full IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title_fullStr IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title_full_unstemmed IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title_short IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation
title_sort il4i1 augments cns remyelination and axonal protection by modulating t cell driven inflammation
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5382940/
https://www.ncbi.nlm.nih.gov/pubmed/27797811
http://dx.doi.org/10.1093/brain/aww254
work_keys_str_mv AT psachouliakonstantina il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT chamberlainkellya il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT heodongeun il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT davisstephaniee il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT paskusjeremiahd il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT nanescusoniae il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT dupreejeffreyl il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT wynnthomasa il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation
AT huangjeffreyk il4i1augmentscnsremyelinationandaxonalprotectionbymodulatingtcelldriveninflammation