Cargando…

Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats

BACKGROUND: Status epilepticus (SE) can cause neuronal cell death and impaired behavioral function. Acute exposure to potent acetylcholinesterase inhibitors such as soman (GD) can cause prolonged SE activity, micro-hemorrhage and cell death in the hippocampus, thalamus and piriform cortex. Neuroinfl...

Descripción completa

Detalles Bibliográficos
Autores principales: Johnson, Erik A., Guignet, Michelle A., Dao, Thuy L., Hamilton, Tracey A., Kan, Robert K.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4510912/
https://www.ncbi.nlm.nih.gov/pubmed/26203299
http://dx.doi.org/10.1186/s12950-015-0089-9
_version_ 1782382262980968448
author Johnson, Erik A.
Guignet, Michelle A.
Dao, Thuy L.
Hamilton, Tracey A.
Kan, Robert K.
author_facet Johnson, Erik A.
Guignet, Michelle A.
Dao, Thuy L.
Hamilton, Tracey A.
Kan, Robert K.
author_sort Johnson, Erik A.
collection PubMed
description BACKGROUND: Status epilepticus (SE) can cause neuronal cell death and impaired behavioral function. Acute exposure to potent acetylcholinesterase inhibitors such as soman (GD) can cause prolonged SE activity, micro-hemorrhage and cell death in the hippocampus, thalamus and piriform cortex. Neuroinflammation is a prominent feature of brain injury with upregulation of multiple pro-inflammatory cytokines including those of the IL-1 family. The highly pleiotropic pro-inflammatory cytokine interleukin-18 (IL-18) belongs to the IL-1 family of cytokines and can propagate neuroinflammation by promoting immune cell infiltration, leukocyte and lymphocyte activation, and angiogenesis and helps facilitate the transition from the innate to the adaptive immune response. The purpose of this study is to characterize the regional and temporal expression of IL −18 and related factors in the brain following SE in a rat GD seizure model followed by localization of IL-18 to specific cell types. METHODS: The protein levels of IL-18, vascular endothelial growth factor and interferon gamma was quantified in the lysates of injured brain regions up to 72 h following GD-induced SE onset using bead multiplex immunoassays. IL-18 was localized to various cell types using immunohistochemistry and transmission electron microscopy. In addition, macrophage appearance scoring and T-cell quantification was determined using immunohistochemistry. Micro-hemorrhages were identified using hematoxylin and eosin staining of brain sections. RESULTS: Significant increases in IL-18 occurred in the piriform cortex, hippocampus and thalamus following SE. IL-18 was primarily expressed by endothelial cells and astrocytes associated with the damaged neurovascular unit. The increase in IL-18 was not related to macrophage accumulation, neutrophil infiltration or T-cell appearance in the injured tissue. CONCLUSIONS: These data show that IL-18 is significantly upregulated following GD-induced SE and localized primarily to endothelial cells in damaged brain vasculature. IL-18 upregulation occurred following leukocyte/lymphocyte infiltration and in the absence of other IL-18-related cytokines, suggesting another function, potentially for angiogenesis related to GD-induced micro-hemorrhage formation. Further studies at more chronic time points may help to elucidate this function.
format Online
Article
Text
id pubmed-4510912
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-45109122015-07-23 Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats Johnson, Erik A. Guignet, Michelle A. Dao, Thuy L. Hamilton, Tracey A. Kan, Robert K. J Inflamm (Lond) Research BACKGROUND: Status epilepticus (SE) can cause neuronal cell death and impaired behavioral function. Acute exposure to potent acetylcholinesterase inhibitors such as soman (GD) can cause prolonged SE activity, micro-hemorrhage and cell death in the hippocampus, thalamus and piriform cortex. Neuroinflammation is a prominent feature of brain injury with upregulation of multiple pro-inflammatory cytokines including those of the IL-1 family. The highly pleiotropic pro-inflammatory cytokine interleukin-18 (IL-18) belongs to the IL-1 family of cytokines and can propagate neuroinflammation by promoting immune cell infiltration, leukocyte and lymphocyte activation, and angiogenesis and helps facilitate the transition from the innate to the adaptive immune response. The purpose of this study is to characterize the regional and temporal expression of IL −18 and related factors in the brain following SE in a rat GD seizure model followed by localization of IL-18 to specific cell types. METHODS: The protein levels of IL-18, vascular endothelial growth factor and interferon gamma was quantified in the lysates of injured brain regions up to 72 h following GD-induced SE onset using bead multiplex immunoassays. IL-18 was localized to various cell types using immunohistochemistry and transmission electron microscopy. In addition, macrophage appearance scoring and T-cell quantification was determined using immunohistochemistry. Micro-hemorrhages were identified using hematoxylin and eosin staining of brain sections. RESULTS: Significant increases in IL-18 occurred in the piriform cortex, hippocampus and thalamus following SE. IL-18 was primarily expressed by endothelial cells and astrocytes associated with the damaged neurovascular unit. The increase in IL-18 was not related to macrophage accumulation, neutrophil infiltration or T-cell appearance in the injured tissue. CONCLUSIONS: These data show that IL-18 is significantly upregulated following GD-induced SE and localized primarily to endothelial cells in damaged brain vasculature. IL-18 upregulation occurred following leukocyte/lymphocyte infiltration and in the absence of other IL-18-related cytokines, suggesting another function, potentially for angiogenesis related to GD-induced micro-hemorrhage formation. Further studies at more chronic time points may help to elucidate this function. BioMed Central 2015-07-22 /pmc/articles/PMC4510912/ /pubmed/26203299 http://dx.doi.org/10.1186/s12950-015-0089-9 Text en © Johnson et al. 2015 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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
Johnson, Erik A.
Guignet, Michelle A.
Dao, Thuy L.
Hamilton, Tracey A.
Kan, Robert K.
Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title_full Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title_fullStr Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title_full_unstemmed Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title_short Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
title_sort interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4510912/
https://www.ncbi.nlm.nih.gov/pubmed/26203299
http://dx.doi.org/10.1186/s12950-015-0089-9
work_keys_str_mv AT johnsonerika interleukin18expressionincreasesinresponsetoneurovasculardamagefollowingsomaninducedstatusepilepticusinrats
AT guignetmichellea interleukin18expressionincreasesinresponsetoneurovasculardamagefollowingsomaninducedstatusepilepticusinrats
AT daothuyl interleukin18expressionincreasesinresponsetoneurovasculardamagefollowingsomaninducedstatusepilepticusinrats
AT hamiltontraceya interleukin18expressionincreasesinresponsetoneurovasculardamagefollowingsomaninducedstatusepilepticusinrats
AT kanrobertk interleukin18expressionincreasesinresponsetoneurovasculardamagefollowingsomaninducedstatusepilepticusinrats