Cargando…

Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats

Studies have shown that exogenous thiamine (THI) supplementation can alleviate inflammation and promote rumen epithelial development in goats and cows. This research aimed to evaluate the effect of THI supplementation on LPS-induced inflammation and energy metabolic dysregulation in RECs of goats. C...

Descripción completa

Detalles Bibliográficos
Autores principales: Ma, Yi, Elmhadi, Mawda, Wang, Chao, Li, Zelin, Zhang, Hao, He, Banglin, Zhao, Xiujuan, Zhang, Zhenbin, Wang, Hongrong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9598694/
https://www.ncbi.nlm.nih.gov/pubmed/36290775
http://dx.doi.org/10.3390/antiox11102048
_version_ 1784816413942218752
author Ma, Yi
Elmhadi, Mawda
Wang, Chao
Li, Zelin
Zhang, Hao
He, Banglin
Zhao, Xiujuan
Zhang, Zhenbin
Wang, Hongrong
author_facet Ma, Yi
Elmhadi, Mawda
Wang, Chao
Li, Zelin
Zhang, Hao
He, Banglin
Zhao, Xiujuan
Zhang, Zhenbin
Wang, Hongrong
author_sort Ma, Yi
collection PubMed
description Studies have shown that exogenous thiamine (THI) supplementation can alleviate inflammation and promote rumen epithelial development in goats and cows. This research aimed to evaluate the effect of THI supplementation on LPS-induced inflammation and energy metabolic dysregulation in RECs of goats. Cells were stimulated with either 5 μg/mL THI for 18 h (THI group) or with 5 μg/mL LPS for 6 h (LPS group). The CON group was stimulated with DMEM/F-12 medium without THI for 18 h. The LPTH group was pretreated with THI for 18 h, followed by LPS stimulation for 6 h. THI supplementation decreased the ROS content (p < 0.05), as well as the ratios of phosphorylated (p)-p65 to p65 (p < 0.05) and p-AMPKα to AMPKα (p < 0.05). Interestingly, when the p38 gene was overexpressed in the LPTH group, the ratio of p-p65 to p65 and p-AMPKα to AMPKα proteins significantly increased, and ATP content decreased (p < 0.05). Our results suggest that THI possesses anti-inflammatory and metabolic-modulatory effects in RECs. The mechanism is largely related to the suppression of the NF-κB/p38 MAPK/AMPK signaling pathway. Additionally, we also revealed that THI supplementation can inhibit LPS-induced oxidative damage and apoptosis to protect mitochondrial function in RECs.
format Online
Article
Text
id pubmed-9598694
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-95986942022-10-27 Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats Ma, Yi Elmhadi, Mawda Wang, Chao Li, Zelin Zhang, Hao He, Banglin Zhao, Xiujuan Zhang, Zhenbin Wang, Hongrong Antioxidants (Basel) Article Studies have shown that exogenous thiamine (THI) supplementation can alleviate inflammation and promote rumen epithelial development in goats and cows. This research aimed to evaluate the effect of THI supplementation on LPS-induced inflammation and energy metabolic dysregulation in RECs of goats. Cells were stimulated with either 5 μg/mL THI for 18 h (THI group) or with 5 μg/mL LPS for 6 h (LPS group). The CON group was stimulated with DMEM/F-12 medium without THI for 18 h. The LPTH group was pretreated with THI for 18 h, followed by LPS stimulation for 6 h. THI supplementation decreased the ROS content (p < 0.05), as well as the ratios of phosphorylated (p)-p65 to p65 (p < 0.05) and p-AMPKα to AMPKα (p < 0.05). Interestingly, when the p38 gene was overexpressed in the LPTH group, the ratio of p-p65 to p65 and p-AMPKα to AMPKα proteins significantly increased, and ATP content decreased (p < 0.05). Our results suggest that THI possesses anti-inflammatory and metabolic-modulatory effects in RECs. The mechanism is largely related to the suppression of the NF-κB/p38 MAPK/AMPK signaling pathway. Additionally, we also revealed that THI supplementation can inhibit LPS-induced oxidative damage and apoptosis to protect mitochondrial function in RECs. MDPI 2022-10-18 /pmc/articles/PMC9598694/ /pubmed/36290775 http://dx.doi.org/10.3390/antiox11102048 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/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 (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Ma, Yi
Elmhadi, Mawda
Wang, Chao
Li, Zelin
Zhang, Hao
He, Banglin
Zhao, Xiujuan
Zhang, Zhenbin
Wang, Hongrong
Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title_full Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title_fullStr Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title_full_unstemmed Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title_short Thiamine Supplementation Alleviates Lipopolysaccharide-Triggered Adaptive Inflammatory Response and Modulates Energy State via Suppression of NFκB/p38 MAPK/AMPK Signaling in Rumen Epithelial Cells of Goats
title_sort thiamine supplementation alleviates lipopolysaccharide-triggered adaptive inflammatory response and modulates energy state via suppression of nfκb/p38 mapk/ampk signaling in rumen epithelial cells of goats
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9598694/
https://www.ncbi.nlm.nih.gov/pubmed/36290775
http://dx.doi.org/10.3390/antiox11102048
work_keys_str_mv AT mayi thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT elmhadimawda thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT wangchao thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT lizelin thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT zhanghao thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT hebanglin thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT zhaoxiujuan thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT zhangzhenbin thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats
AT wanghongrong thiaminesupplementationalleviateslipopolysaccharidetriggeredadaptiveinflammatoryresponseandmodulatesenergystateviasuppressionofnfkbp38mapkampksignalinginrumenepithelialcellsofgoats