Cargando…

RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis

N(6)-methyladenosine (m(6)A), the most prevalent mRNA modification, has an important function in diverse biological processes. However, the involvement of m(6)A in allergic asthma and macrophage homeostasis remains largely unknown. Here we show that m(6)A methyltransferases METTL3 is expressed at a...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Xiao, Liu, Lijuan, Huang, Saihua, Xiao, Wenfeng, Gao, Yajing, Zhou, Weitao, Zhang, Caiyan, Zheng, Hongmei, Yang, Lan, Xie, Xueru, Liang, Qiuyan, Tu, Zikun, Yu, Hongmiao, Fu, Jinrong, Wang, Libo, Zhang, Xiaobo, Qian, Liling, Zhou, Yufeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10643624/
https://www.ncbi.nlm.nih.gov/pubmed/37957139
http://dx.doi.org/10.1038/s41467-023-43219-w
_version_ 1785147140973002752
author Han, Xiao
Liu, Lijuan
Huang, Saihua
Xiao, Wenfeng
Gao, Yajing
Zhou, Weitao
Zhang, Caiyan
Zheng, Hongmei
Yang, Lan
Xie, Xueru
Liang, Qiuyan
Tu, Zikun
Yu, Hongmiao
Fu, Jinrong
Wang, Libo
Zhang, Xiaobo
Qian, Liling
Zhou, Yufeng
author_facet Han, Xiao
Liu, Lijuan
Huang, Saihua
Xiao, Wenfeng
Gao, Yajing
Zhou, Weitao
Zhang, Caiyan
Zheng, Hongmei
Yang, Lan
Xie, Xueru
Liang, Qiuyan
Tu, Zikun
Yu, Hongmiao
Fu, Jinrong
Wang, Libo
Zhang, Xiaobo
Qian, Liling
Zhou, Yufeng
author_sort Han, Xiao
collection PubMed
description N(6)-methyladenosine (m(6)A), the most prevalent mRNA modification, has an important function in diverse biological processes. However, the involvement of m(6)A in allergic asthma and macrophage homeostasis remains largely unknown. Here we show that m(6)A methyltransferases METTL3 is expressed at a low level in monocyte-derived macrophages from childhood allergic asthma patients. Conditional knockout of Mettl3 in myeloid cells enhances Th2 cell response and aggravates allergic airway inflammation by facilitating M2 macrophage activation. Loss and gain functional studies confirm that METTL3 suppresses M2 macrophage activation partly through PI3K/AKT and JAK/STAT6 signaling. Mechanistically, m(6)A-sequencing shows that loss of METTL3 impairs the m(6)A-YTHDF3-dependent degradation of PTX3 mRNA, while higher PTX3 expression positively correlates with asthma severity through promoting M2 macrophage activation. Furthermore, the METTL3/YTHDF3-m(6)A/PTX3 interactions contribute to autophagy maturation in macrophages by modulating STX17 expression. Collectively, this study highlights the function of m(6)A in regulating macrophage homeostasis and identifies potential targets in controlling allergic asthma.
format Online
Article
Text
id pubmed-10643624
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-106436242023-11-13 RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis Han, Xiao Liu, Lijuan Huang, Saihua Xiao, Wenfeng Gao, Yajing Zhou, Weitao Zhang, Caiyan Zheng, Hongmei Yang, Lan Xie, Xueru Liang, Qiuyan Tu, Zikun Yu, Hongmiao Fu, Jinrong Wang, Libo Zhang, Xiaobo Qian, Liling Zhou, Yufeng Nat Commun Article N(6)-methyladenosine (m(6)A), the most prevalent mRNA modification, has an important function in diverse biological processes. However, the involvement of m(6)A in allergic asthma and macrophage homeostasis remains largely unknown. Here we show that m(6)A methyltransferases METTL3 is expressed at a low level in monocyte-derived macrophages from childhood allergic asthma patients. Conditional knockout of Mettl3 in myeloid cells enhances Th2 cell response and aggravates allergic airway inflammation by facilitating M2 macrophage activation. Loss and gain functional studies confirm that METTL3 suppresses M2 macrophage activation partly through PI3K/AKT and JAK/STAT6 signaling. Mechanistically, m(6)A-sequencing shows that loss of METTL3 impairs the m(6)A-YTHDF3-dependent degradation of PTX3 mRNA, while higher PTX3 expression positively correlates with asthma severity through promoting M2 macrophage activation. Furthermore, the METTL3/YTHDF3-m(6)A/PTX3 interactions contribute to autophagy maturation in macrophages by modulating STX17 expression. Collectively, this study highlights the function of m(6)A in regulating macrophage homeostasis and identifies potential targets in controlling allergic asthma. Nature Publishing Group UK 2023-11-13 /pmc/articles/PMC10643624/ /pubmed/37957139 http://dx.doi.org/10.1038/s41467-023-43219-w Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Han, Xiao
Liu, Lijuan
Huang, Saihua
Xiao, Wenfeng
Gao, Yajing
Zhou, Weitao
Zhang, Caiyan
Zheng, Hongmei
Yang, Lan
Xie, Xueru
Liang, Qiuyan
Tu, Zikun
Yu, Hongmiao
Fu, Jinrong
Wang, Libo
Zhang, Xiaobo
Qian, Liling
Zhou, Yufeng
RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title_full RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title_fullStr RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title_full_unstemmed RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title_short RNA m(6)A methylation modulates airway inflammation in allergic asthma via PTX3-dependent macrophage homeostasis
title_sort rna m(6)a methylation modulates airway inflammation in allergic asthma via ptx3-dependent macrophage homeostasis
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10643624/
https://www.ncbi.nlm.nih.gov/pubmed/37957139
http://dx.doi.org/10.1038/s41467-023-43219-w
work_keys_str_mv AT hanxiao rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT liulijuan rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT huangsaihua rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT xiaowenfeng rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT gaoyajing rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT zhouweitao rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT zhangcaiyan rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT zhenghongmei rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT yanglan rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT xiexueru rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT liangqiuyan rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT tuzikun rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT yuhongmiao rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT fujinrong rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT wanglibo rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT zhangxiaobo rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT qianliling rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis
AT zhouyufeng rnam6amethylationmodulatesairwayinflammationinallergicasthmaviaptx3dependentmacrophagehomeostasis