Cargando…

Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain

BACKGROUND: Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic...

Descripción completa

Detalles Bibliográficos
Autores principales: Doyen, Pierre J., Vergouts, Maxime, Pochet, Amandine, Desmet, Nathalie, van Neerven, Sabien, Brook, Gary, Hermans, Emmanuel
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5658970/
https://www.ncbi.nlm.nih.gov/pubmed/29078779
http://dx.doi.org/10.1186/s12974-017-0971-x
_version_ 1783274087694467072
author Doyen, Pierre J.
Vergouts, Maxime
Pochet, Amandine
Desmet, Nathalie
van Neerven, Sabien
Brook, Gary
Hermans, Emmanuel
author_facet Doyen, Pierre J.
Vergouts, Maxime
Pochet, Amandine
Desmet, Nathalie
van Neerven, Sabien
Brook, Gary
Hermans, Emmanuel
author_sort Doyen, Pierre J.
collection PubMed
description BACKGROUND: Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic pain conditions may rely on alterations in RGS function. The expression and the regulation of RGS in astrocytes is poorly documented, and we herein hypothesized that neuroinflammation which is commonly observed in neuropathic pain could influence RGS expression in astrocytes. METHODS: In a validated model of neuropathic pain, the spared nerve injury (SNI), the regulation of RGS2, RGS3, RGS4, and RGS7 messenger RNA (mRNA) was examined up to 3 weeks after the lesion. Changes in the expression of the same RGS were also studied in cultured astrocytes exposed to defined activation protocols or to inflammatory cytokines. RESULTS: We evidenced a differential regulation of these RGS in the lumbar spinal cord of animals undergoing SNI. In particular, RGS3 appeared upregulated at early stages after the lesion whereas expression of RGS2 and RGS4 was decreased at later stages. Decrease in RGS7 expression was already observed after 3 days and outlasted until 21 days after the lesion. In cultured astrocytes, we observed that changes in the culture conditions distinctly influenced the constitutive expression of these RGS. Also, brief exposures (4 to 8 h) to either interleukin-1β, interleukin-6, or tumor necrosis factor α caused rapid changes in the mRNA levels of the RGS, which however did not strictly recapitulate the regulations observed in the spinal cord of lesioned animals. Longer exposure (48 h) to inflammatory cytokines barely influenced RGS expression, confirming the rapid but transient regulation of these cell signaling modulators. CONCLUSION: Changes in the environment of astrocytes mimicking the inflammation observed in the model of neuropathic pain can affect RGS expression. Considering the role of astrocytes in the onset and progression of neuropathic pain, we propose that the inflammation-mediated modulation of RGS in astrocytes constitutes an adaptive mechanism in a context of neuroinflammation and may participate in the regulation of nociception.
format Online
Article
Text
id pubmed-5658970
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-56589702017-11-01 Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain Doyen, Pierre J. Vergouts, Maxime Pochet, Amandine Desmet, Nathalie van Neerven, Sabien Brook, Gary Hermans, Emmanuel J Neuroinflammation Research BACKGROUND: Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic pain conditions may rely on alterations in RGS function. The expression and the regulation of RGS in astrocytes is poorly documented, and we herein hypothesized that neuroinflammation which is commonly observed in neuropathic pain could influence RGS expression in astrocytes. METHODS: In a validated model of neuropathic pain, the spared nerve injury (SNI), the regulation of RGS2, RGS3, RGS4, and RGS7 messenger RNA (mRNA) was examined up to 3 weeks after the lesion. Changes in the expression of the same RGS were also studied in cultured astrocytes exposed to defined activation protocols or to inflammatory cytokines. RESULTS: We evidenced a differential regulation of these RGS in the lumbar spinal cord of animals undergoing SNI. In particular, RGS3 appeared upregulated at early stages after the lesion whereas expression of RGS2 and RGS4 was decreased at later stages. Decrease in RGS7 expression was already observed after 3 days and outlasted until 21 days after the lesion. In cultured astrocytes, we observed that changes in the culture conditions distinctly influenced the constitutive expression of these RGS. Also, brief exposures (4 to 8 h) to either interleukin-1β, interleukin-6, or tumor necrosis factor α caused rapid changes in the mRNA levels of the RGS, which however did not strictly recapitulate the regulations observed in the spinal cord of lesioned animals. Longer exposure (48 h) to inflammatory cytokines barely influenced RGS expression, confirming the rapid but transient regulation of these cell signaling modulators. CONCLUSION: Changes in the environment of astrocytes mimicking the inflammation observed in the model of neuropathic pain can affect RGS expression. Considering the role of astrocytes in the onset and progression of neuropathic pain, we propose that the inflammation-mediated modulation of RGS in astrocytes constitutes an adaptive mechanism in a context of neuroinflammation and may participate in the regulation of nociception. BioMed Central 2017-10-27 /pmc/articles/PMC5658970/ /pubmed/29078779 http://dx.doi.org/10.1186/s12974-017-0971-x Text en © The Author(s). 2017 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Doyen, Pierre J.
Vergouts, Maxime
Pochet, Amandine
Desmet, Nathalie
van Neerven, Sabien
Brook, Gary
Hermans, Emmanuel
Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title_full Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title_fullStr Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title_full_unstemmed Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title_short Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain
title_sort inflammation-associated regulation of rgs in astrocytes and putative implication in neuropathic pain
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5658970/
https://www.ncbi.nlm.nih.gov/pubmed/29078779
http://dx.doi.org/10.1186/s12974-017-0971-x
work_keys_str_mv AT doyenpierrej inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT vergoutsmaxime inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT pochetamandine inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT desmetnathalie inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT vanneervensabien inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT brookgary inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain
AT hermansemmanuel inflammationassociatedregulationofrgsinastrocytesandputativeimplicationinneuropathicpain