Cargando…

Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation

Allergic diseases are associated with central and peripheral nervous system diseases such as autism spectrum disorders and eosinophilic granulomatosis with polyangiitis, which frequently causes mononeuritis multiplex. Thus, it is possible that patients with an atopic constitution might develop multi...

Descripción completa

Detalles Bibliográficos
Autores principales: Fujii, Takayuki, Yamasaki, Ryo, Kira, Jun-ichi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6928142/
https://www.ncbi.nlm.nih.gov/pubmed/31920952
http://dx.doi.org/10.3389/fneur.2019.01337
_version_ 1783482419670679552
author Fujii, Takayuki
Yamasaki, Ryo
Kira, Jun-ichi
author_facet Fujii, Takayuki
Yamasaki, Ryo
Kira, Jun-ichi
author_sort Fujii, Takayuki
collection PubMed
description Allergic diseases are associated with central and peripheral nervous system diseases such as autism spectrum disorders and eosinophilic granulomatosis with polyangiitis, which frequently causes mononeuritis multiplex. Thus, it is possible that patients with an atopic constitution might develop multifocal inflammation in central and peripheral nervous system tissues. In a previous study in Japan, we reported a rare form of myelitis with persistent neuropathic pain (NeP) in patients with allergic disorders. However, the underlying mechanism of allergic inflammation-related NeP remains to be elucidated. First, we analyzed the effect of allergic inflammation on the nociceptive system in the spinal cord. Mice with atopy showed microglial and astroglial activation in the spinal cord and tactile allodynia. In a microarray analysis of isolated microglia from the spinal cord, endothelin receptor type B (EDNRB) was the most upregulated cell surface receptor in mice with atopy. Immunohistochemical analysis demonstrated EDNRB expression was upregulated in microglia and astroglia. The EDNRB antagonist BQ788 abolished glial activation and allodynia. These findings indicated that allergic inflammation induced widespread glial activation through the EDNRB pathway and NeP. Second, we investigated whether autoantibody-mediated pathogenesis underlies allergic inflammation-related NeP. We detected specific autoantibodies to small dorsal root ganglion (DRG) neurons and their nerve terminals in the dorsal horns of NeP patients with allergic disorders. An analysis of IgG subclasses revealed a predominance of IgG2. These autoantibodies were mostly colocalized with isolectin B4- and P2X3-positive unmyelinated C-fiber type small DRG neurons. By contrast, immunostaining for S100β, a myelinated DRG neuron marker, showed no colocalization with patient IgG. Immunoprecipitation and liquid chromatography-tandem mass spectrometry identified plexin D1 as a target autoantigen. Patients with anti-plexin D1 antibodies often present with burning pain and thermal hyperalgesia. Immunotherapies, including plasma exchange, are effective for NeP management. Therefore, anti-plexin D1 antibodies may be pathogenic for immune-mediated NeP, especially under allergic inflammation conditions. Thus, allergic inflammation may induce NeP through glial inflammation in the spinal cord and the anti-plexin D1 antibody-mediated impairment of small DRG neurons.
format Online
Article
Text
id pubmed-6928142
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-69281422020-01-09 Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation Fujii, Takayuki Yamasaki, Ryo Kira, Jun-ichi Front Neurol Neurology Allergic diseases are associated with central and peripheral nervous system diseases such as autism spectrum disorders and eosinophilic granulomatosis with polyangiitis, which frequently causes mononeuritis multiplex. Thus, it is possible that patients with an atopic constitution might develop multifocal inflammation in central and peripheral nervous system tissues. In a previous study in Japan, we reported a rare form of myelitis with persistent neuropathic pain (NeP) in patients with allergic disorders. However, the underlying mechanism of allergic inflammation-related NeP remains to be elucidated. First, we analyzed the effect of allergic inflammation on the nociceptive system in the spinal cord. Mice with atopy showed microglial and astroglial activation in the spinal cord and tactile allodynia. In a microarray analysis of isolated microglia from the spinal cord, endothelin receptor type B (EDNRB) was the most upregulated cell surface receptor in mice with atopy. Immunohistochemical analysis demonstrated EDNRB expression was upregulated in microglia and astroglia. The EDNRB antagonist BQ788 abolished glial activation and allodynia. These findings indicated that allergic inflammation induced widespread glial activation through the EDNRB pathway and NeP. Second, we investigated whether autoantibody-mediated pathogenesis underlies allergic inflammation-related NeP. We detected specific autoantibodies to small dorsal root ganglion (DRG) neurons and their nerve terminals in the dorsal horns of NeP patients with allergic disorders. An analysis of IgG subclasses revealed a predominance of IgG2. These autoantibodies were mostly colocalized with isolectin B4- and P2X3-positive unmyelinated C-fiber type small DRG neurons. By contrast, immunostaining for S100β, a myelinated DRG neuron marker, showed no colocalization with patient IgG. Immunoprecipitation and liquid chromatography-tandem mass spectrometry identified plexin D1 as a target autoantigen. Patients with anti-plexin D1 antibodies often present with burning pain and thermal hyperalgesia. Immunotherapies, including plasma exchange, are effective for NeP management. Therefore, anti-plexin D1 antibodies may be pathogenic for immune-mediated NeP, especially under allergic inflammation conditions. Thus, allergic inflammation may induce NeP through glial inflammation in the spinal cord and the anti-plexin D1 antibody-mediated impairment of small DRG neurons. Frontiers Media S.A. 2019-12-17 /pmc/articles/PMC6928142/ /pubmed/31920952 http://dx.doi.org/10.3389/fneur.2019.01337 Text en Copyright © 2019 Fujii, Yamasaki and Kira. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Neurology
Fujii, Takayuki
Yamasaki, Ryo
Kira, Jun-ichi
Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title_full Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title_fullStr Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title_full_unstemmed Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title_short Novel Neuropathic Pain Mechanisms Associated With Allergic Inflammation
title_sort novel neuropathic pain mechanisms associated with allergic inflammation
topic Neurology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6928142/
https://www.ncbi.nlm.nih.gov/pubmed/31920952
http://dx.doi.org/10.3389/fneur.2019.01337
work_keys_str_mv AT fujiitakayuki novelneuropathicpainmechanismsassociatedwithallergicinflammation
AT yamasakiryo novelneuropathicpainmechanismsassociatedwithallergicinflammation
AT kirajunichi novelneuropathicpainmechanismsassociatedwithallergicinflammation