Cargando…

Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway

The imbalance between Th17 and Treg cells substantially contributes to the intestinal immune disturbance and subsequent tissue injury in ulcerative colitis. The triterpenoid-rich fraction of Centella asiatica was able to ameliorate dextran sulfate sodium-induced colitis in mice. Here we explored its...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Xiaotian, Wang, Yuhui, Wei, Zhifeng, Wei, Wenhui, Zhao, Peng, Tong, Bei, Xia, Yufeng, Dai, Yue
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5386545/
https://www.ncbi.nlm.nih.gov/pubmed/28358365
http://dx.doi.org/10.1038/cddis.2017.150
_version_ 1782520786453528576
author Xu, Xiaotian
Wang, Yuhui
Wei, Zhifeng
Wei, Wenhui
Zhao, Peng
Tong, Bei
Xia, Yufeng
Dai, Yue
author_facet Xu, Xiaotian
Wang, Yuhui
Wei, Zhifeng
Wei, Wenhui
Zhao, Peng
Tong, Bei
Xia, Yufeng
Dai, Yue
author_sort Xu, Xiaotian
collection PubMed
description The imbalance between Th17 and Treg cells substantially contributes to the intestinal immune disturbance and subsequent tissue injury in ulcerative colitis. The triterpenoid-rich fraction of Centella asiatica was able to ameliorate dextran sulfate sodium-induced colitis in mice. Here we explored its active ingredient and underlying mechanism with a focus on restoring the Th17/Treg balance. The four main triterpenoids occurring in C. asiatica were shown to attenuate colitis in mice by oral administration. The most effective ingredient madecassoside lost anti-colitis effect when applied topically in the colon, and madecassic acid was recognized to be the active form of madecassoside. Oral administration of madecassic acid decreased the percentage of Th17 cells and downregulated the expression of RORγt, IL-17A, IL-17F, IL-21 and IL-22 and increased the percentage of Treg cells and the expression of Foxp3 and IL-10 in the colons of mice with colitis, but it did not affect Th1 and Th2 cells. Under Th17-polarizing conditions, madecassic acid downregulated ACC1 expression and enhanced the shift of Th17 cells toward Treg cells, but it did not affect the differentiation of Treg cells under Treg-polarizing conditions. Both compound C and AMPK siRNA inhibited the madecassic acid-mediated downregulation of ACC1 expression and shift of Th17 cells to Treg cells under Th17-polarizing conditions. GW9662, T0070907 and PPARγ siRNA blocked the effect of madecassic acid on AMPK activation, ACC1 expression and shift of Th17 cells to Treg cells. Furthermore, madecassic acid was identified as a PPARγ agonist, as it promoted PPARγ transactivation. The correlation between activation of PPARγ and AMPK, downregulation of ACC1 expression, restoration of Th17/Treg balance and attenuation of colitis by madecassic acid was validated in mice with DSS-induced colitis. In conclusion, madecassic acid was the active form of madecassoside in ameliorating colitis by restoring the Th17/Treg balance via regulating the PPARγ/AMPK/ACC1 pathway.
format Online
Article
Text
id pubmed-5386545
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-53865452017-04-27 Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway Xu, Xiaotian Wang, Yuhui Wei, Zhifeng Wei, Wenhui Zhao, Peng Tong, Bei Xia, Yufeng Dai, Yue Cell Death Dis Original Article The imbalance between Th17 and Treg cells substantially contributes to the intestinal immune disturbance and subsequent tissue injury in ulcerative colitis. The triterpenoid-rich fraction of Centella asiatica was able to ameliorate dextran sulfate sodium-induced colitis in mice. Here we explored its active ingredient and underlying mechanism with a focus on restoring the Th17/Treg balance. The four main triterpenoids occurring in C. asiatica were shown to attenuate colitis in mice by oral administration. The most effective ingredient madecassoside lost anti-colitis effect when applied topically in the colon, and madecassic acid was recognized to be the active form of madecassoside. Oral administration of madecassic acid decreased the percentage of Th17 cells and downregulated the expression of RORγt, IL-17A, IL-17F, IL-21 and IL-22 and increased the percentage of Treg cells and the expression of Foxp3 and IL-10 in the colons of mice with colitis, but it did not affect Th1 and Th2 cells. Under Th17-polarizing conditions, madecassic acid downregulated ACC1 expression and enhanced the shift of Th17 cells toward Treg cells, but it did not affect the differentiation of Treg cells under Treg-polarizing conditions. Both compound C and AMPK siRNA inhibited the madecassic acid-mediated downregulation of ACC1 expression and shift of Th17 cells to Treg cells under Th17-polarizing conditions. GW9662, T0070907 and PPARγ siRNA blocked the effect of madecassic acid on AMPK activation, ACC1 expression and shift of Th17 cells to Treg cells. Furthermore, madecassic acid was identified as a PPARγ agonist, as it promoted PPARγ transactivation. The correlation between activation of PPARγ and AMPK, downregulation of ACC1 expression, restoration of Th17/Treg balance and attenuation of colitis by madecassic acid was validated in mice with DSS-induced colitis. In conclusion, madecassic acid was the active form of madecassoside in ameliorating colitis by restoring the Th17/Treg balance via regulating the PPARγ/AMPK/ACC1 pathway. Nature Publishing Group 2017-03 2017-03-30 /pmc/articles/PMC5386545/ /pubmed/28358365 http://dx.doi.org/10.1038/cddis.2017.150 Text en Copyright © 2017 The Author(s) http://creativecommons.org/licenses/by/4.0/ Cell Death and Disease is an open-access journal published by Nature Publishing Group. This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article's Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Original Article
Xu, Xiaotian
Wang, Yuhui
Wei, Zhifeng
Wei, Wenhui
Zhao, Peng
Tong, Bei
Xia, Yufeng
Dai, Yue
Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title_full Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title_fullStr Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title_full_unstemmed Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title_short Madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of Th17 toward Treg cells via the PPARγ/AMPK/ACC1 pathway
title_sort madecassic acid, the contributor to the anti-colitis effect of madecassoside, enhances the shift of th17 toward treg cells via the pparγ/ampk/acc1 pathway
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5386545/
https://www.ncbi.nlm.nih.gov/pubmed/28358365
http://dx.doi.org/10.1038/cddis.2017.150
work_keys_str_mv AT xuxiaotian madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT wangyuhui madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT weizhifeng madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT weiwenhui madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT zhaopeng madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT tongbei madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT xiayufeng madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway
AT daiyue madecassicacidthecontributortotheanticolitiseffectofmadecassosideenhancestheshiftofth17towardtregcellsviatheppargampkacc1pathway