Cargando…

Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model

Hyperbaric oxygen (HBO) treatment has been proven to decrease neuroinflammation in rats. This study aimed to determine the potential mechanism underlying the anti-inflammatory effects of HBO treatment on burn-induced neuroinflammation in rats. Thirty-six adult male Sprague-Dawley (SD) rats were rand...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Zong-Sheng, Lo, Jing-Jou, Wu, Sheng-Hua, Wang, Chau-Zen, Chen, Rong-Fu, Lee, Su-Shin, Chai, Chee-Yin, Huang, Shu-Hung
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6121430/
https://www.ncbi.nlm.nih.gov/pubmed/30060489
http://dx.doi.org/10.3390/ijms19082195
_version_ 1783352466061918208
author Wu, Zong-Sheng
Lo, Jing-Jou
Wu, Sheng-Hua
Wang, Chau-Zen
Chen, Rong-Fu
Lee, Su-Shin
Chai, Chee-Yin
Huang, Shu-Hung
author_facet Wu, Zong-Sheng
Lo, Jing-Jou
Wu, Sheng-Hua
Wang, Chau-Zen
Chen, Rong-Fu
Lee, Su-Shin
Chai, Chee-Yin
Huang, Shu-Hung
author_sort Wu, Zong-Sheng
collection PubMed
description Hyperbaric oxygen (HBO) treatment has been proven to decrease neuroinflammation in rats. This study aimed to determine the potential mechanism underlying the anti-inflammatory effects of HBO treatment on burn-induced neuroinflammation in rats. Thirty-six adult male Sprague-Dawley (SD) rats were randomly assigned to the following six groups (n = 6 per group): (1) sham burn with sham HBO treatment; (2) sham burn with HBO treatment; (3) burn with one-week sham HBO treatment; (4) burn with two-week sham HBO treatment; (5) burn with one-week HBO treatment; and (6) burn with two-week HBO treatment. SD rats that received third-degree burn injury were used as a full-thickness burn injury model. Subsequently, we analyzed the expression of proteins involved in the galectin-3 (Gal-3)-dependent Toll-like receptor-4 (TLR-4) pathway through enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) analysis, and Western blotting. A behavior test was also conducted, which revealed that HBO treatment significantly suppressed mechanical hypersensitivity in the burn with HBO treatment group compared to the burn with sham HBO treatment group (p < 0.05). ELISA results showed that tumor necrosis factor α (TNF-α) and interleukin 1 beta (IL-1β) levels in the dorsal horn of the spinal cord and the skin significantly decreased in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). Western blotting results demonstrated that HBO treatment significantly reduced the expression of Gal-3 and TLR-4 in the dorsal horn of the spinal cord in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). IHC analysis showed that the expression of Gal-3, TLR-4, CD68 and CD45 in the dorsal horn of the spinal cord was significantly lower in the burn with HBO treatment group than in the burn with sham HBO treatment group (p < 0.05), and the expression of CD68 and macrophage migration inhibitory factor (MIF) in the right hind paw skin was significantly lower. The expression of vimentin and fibroblast growth factor in the right hind paw skin was significantly higher after HBO treatment (p < 0.05). This study proved that early HBO treatment relieves neuropathic pain, inhibits the Gal-3-dependent TLR-4 pathway, and suppresses microglia and macrophage activation in a rat model.
format Online
Article
Text
id pubmed-6121430
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-61214302018-09-07 Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model Wu, Zong-Sheng Lo, Jing-Jou Wu, Sheng-Hua Wang, Chau-Zen Chen, Rong-Fu Lee, Su-Shin Chai, Chee-Yin Huang, Shu-Hung Int J Mol Sci Article Hyperbaric oxygen (HBO) treatment has been proven to decrease neuroinflammation in rats. This study aimed to determine the potential mechanism underlying the anti-inflammatory effects of HBO treatment on burn-induced neuroinflammation in rats. Thirty-six adult male Sprague-Dawley (SD) rats were randomly assigned to the following six groups (n = 6 per group): (1) sham burn with sham HBO treatment; (2) sham burn with HBO treatment; (3) burn with one-week sham HBO treatment; (4) burn with two-week sham HBO treatment; (5) burn with one-week HBO treatment; and (6) burn with two-week HBO treatment. SD rats that received third-degree burn injury were used as a full-thickness burn injury model. Subsequently, we analyzed the expression of proteins involved in the galectin-3 (Gal-3)-dependent Toll-like receptor-4 (TLR-4) pathway through enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) analysis, and Western blotting. A behavior test was also conducted, which revealed that HBO treatment significantly suppressed mechanical hypersensitivity in the burn with HBO treatment group compared to the burn with sham HBO treatment group (p < 0.05). ELISA results showed that tumor necrosis factor α (TNF-α) and interleukin 1 beta (IL-1β) levels in the dorsal horn of the spinal cord and the skin significantly decreased in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). Western blotting results demonstrated that HBO treatment significantly reduced the expression of Gal-3 and TLR-4 in the dorsal horn of the spinal cord in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). IHC analysis showed that the expression of Gal-3, TLR-4, CD68 and CD45 in the dorsal horn of the spinal cord was significantly lower in the burn with HBO treatment group than in the burn with sham HBO treatment group (p < 0.05), and the expression of CD68 and macrophage migration inhibitory factor (MIF) in the right hind paw skin was significantly lower. The expression of vimentin and fibroblast growth factor in the right hind paw skin was significantly higher after HBO treatment (p < 0.05). This study proved that early HBO treatment relieves neuropathic pain, inhibits the Gal-3-dependent TLR-4 pathway, and suppresses microglia and macrophage activation in a rat model. MDPI 2018-07-27 /pmc/articles/PMC6121430/ /pubmed/30060489 http://dx.doi.org/10.3390/ijms19082195 Text en © 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Wu, Zong-Sheng
Lo, Jing-Jou
Wu, Sheng-Hua
Wang, Chau-Zen
Chen, Rong-Fu
Lee, Su-Shin
Chai, Chee-Yin
Huang, Shu-Hung
Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title_full Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title_fullStr Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title_full_unstemmed Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title_short Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model
title_sort early hyperbaric oxygen treatment attenuates burn-induced neuroinflammation by inhibiting the galectin-3-dependent toll-like receptor-4 pathway in a rat model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6121430/
https://www.ncbi.nlm.nih.gov/pubmed/30060489
http://dx.doi.org/10.3390/ijms19082195
work_keys_str_mv AT wuzongsheng earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT lojingjou earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT wushenghua earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT wangchauzen earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT chenrongfu earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT leesushin earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT chaicheeyin earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel
AT huangshuhung earlyhyperbaricoxygentreatmentattenuatesburninducedneuroinflammationbyinhibitingthegalectin3dependenttolllikereceptor4pathwayinaratmodel