PM2.5 暴露通过下调 P2Y2 受体/CFTR 通路抑制跨上皮阴离子短路电流

IF 3.2 3区 医学 Q1 MEDICINE, GENERAL & INTERNAL
International Journal of Medical Sciences Pub Date : 2024-07-22 eCollection Date: 2024-01-01 DOI:10.7150/ijms.96777
Xiaolong Liu, Zhangwen Li, Jiajie Shan, Fang Wang, Zhongpeng Li, Shaohua Luo, Jian Wu
{"title":"PM2.5 暴露通过下调 P2Y2 受体/CFTR 通路抑制跨上皮阴离子短路电流","authors":"Xiaolong Liu, Zhangwen Li, Jiajie Shan, Fang Wang, Zhongpeng Li, Shaohua Luo, Jian Wu","doi":"10.7150/ijms.96777","DOIUrl":null,"url":null,"abstract":"<p><p>Fine particulate matter (PM<sub>2.5</sub>) can damage airway epithelial barriers. The anion transport system plays a crucial role in airway epithelial barriers. However, the detrimental effect and mechanism of PM<sub>2.5</sub> on the anion transport system are still unclear. In this study, airway epithelial cells and ovalbumin (OVA)-induced asthmatic mice were used. In transwell model, the adenosine triphosphate (ATP)-induced transepithelial anion short-circuit current (I<sub>sc</sub>) and airway surface liquid (ASL) significantly decreased after PM<sub>2.5</sub> exposure. In addition, PM<sub>2.5</sub> exposure decreased the expression levels of P2Y2R, CFTR and cytoplasmic free-calcium, but ATP can increase the expressions of these proteins. PM<sub>2.5</sub> exposure increased the levels of Th2-related cytokines of bronchoalveolar lavage fluid, lung inflammation, collagen deposition and hyperplasisa of goblet cells. Interestingly, the administration of ATP showed an inhibitory effect on lung inflammation induced by PM<sub>2.5</sub>. Together, our study reveals that PM<sub>2.5</sub> impairs the ATP-induced transepithelial anion I<sub>sc</sub> through downregulating P2Y2R/CFTR pathway, and this process may participate in aggravating airway hyperresponsiveness and airway inflammation. These findings may provide important insights on PM<sub>2.5</sub>-mediated airway epithelial injury.</p>","PeriodicalId":14031,"journal":{"name":"International Journal of Medical Sciences","volume":null,"pages":null},"PeriodicalIF":3.2000,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302563/pdf/","citationCount":"0","resultStr":"{\"title\":\"PM<sub>2.5</sub> Exposure Inhibits Transepithelial Anion Short-circuit Current by Downregulating P2Y2 Receptor/CFTR Pathway.\",\"authors\":\"Xiaolong Liu, Zhangwen Li, Jiajie Shan, Fang Wang, Zhongpeng Li, Shaohua Luo, Jian Wu\",\"doi\":\"10.7150/ijms.96777\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Fine particulate matter (PM<sub>2.5</sub>) can damage airway epithelial barriers. The anion transport system plays a crucial role in airway epithelial barriers. However, the detrimental effect and mechanism of PM<sub>2.5</sub> on the anion transport system are still unclear. In this study, airway epithelial cells and ovalbumin (OVA)-induced asthmatic mice were used. In transwell model, the adenosine triphosphate (ATP)-induced transepithelial anion short-circuit current (I<sub>sc</sub>) and airway surface liquid (ASL) significantly decreased after PM<sub>2.5</sub> exposure. In addition, PM<sub>2.5</sub> exposure decreased the expression levels of P2Y2R, CFTR and cytoplasmic free-calcium, but ATP can increase the expressions of these proteins. PM<sub>2.5</sub> exposure increased the levels of Th2-related cytokines of bronchoalveolar lavage fluid, lung inflammation, collagen deposition and hyperplasisa of goblet cells. Interestingly, the administration of ATP showed an inhibitory effect on lung inflammation induced by PM<sub>2.5</sub>. Together, our study reveals that PM<sub>2.5</sub> impairs the ATP-induced transepithelial anion I<sub>sc</sub> through downregulating P2Y2R/CFTR pathway, and this process may participate in aggravating airway hyperresponsiveness and airway inflammation. These findings may provide important insights on PM<sub>2.5</sub>-mediated airway epithelial injury.</p>\",\"PeriodicalId\":14031,\"journal\":{\"name\":\"International Journal of Medical Sciences\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.2000,\"publicationDate\":\"2024-07-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302563/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"International Journal of Medical Sciences\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.7150/ijms.96777\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, GENERAL & INTERNAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"International Journal of Medical Sciences","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/ijms.96777","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"MEDICINE, GENERAL & INTERNAL","Score":null,"Total":0}
引用次数: 0

摘要

细颗粒物(PM2.5)会损害气道上皮屏障。阴离子转运系统在气道上皮屏障中起着至关重要的作用。然而,PM2.5 对阴离子转运系统的不利影响和机制仍不清楚。本研究使用了气道上皮细胞和卵清蛋白(OVA)诱导的哮喘小鼠。在跨孔模型中,PM2.5暴露后,三磷酸腺苷(ATP)诱导的跨上皮阴离子短路电流(Isc)和气道表面液体(ASL)显著下降。此外,PM2.5 暴露降低了 P2Y2R、CFTR 和细胞质自由钙的表达水平,但 ATP 可增加这些蛋白的表达。暴露于 PM2.5 会增加支气管肺泡灌洗液中 Th2 相关细胞因子的水平、肺部炎症、胶原沉积和小管细胞增生。有趣的是,服用 ATP 对 PM2.5 诱导的肺部炎症有抑制作用。总之,我们的研究揭示了PM2.5通过下调P2Y2R/CFTR途径损害ATP诱导的跨上皮阴离子Isc,这一过程可能参与加剧气道高反应性和气道炎症。这些发现可能会对PM2.5介导的气道上皮损伤提供重要启示。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
PM2.5 Exposure Inhibits Transepithelial Anion Short-circuit Current by Downregulating P2Y2 Receptor/CFTR Pathway.

Fine particulate matter (PM2.5) can damage airway epithelial barriers. The anion transport system plays a crucial role in airway epithelial barriers. However, the detrimental effect and mechanism of PM2.5 on the anion transport system are still unclear. In this study, airway epithelial cells and ovalbumin (OVA)-induced asthmatic mice were used. In transwell model, the adenosine triphosphate (ATP)-induced transepithelial anion short-circuit current (Isc) and airway surface liquid (ASL) significantly decreased after PM2.5 exposure. In addition, PM2.5 exposure decreased the expression levels of P2Y2R, CFTR and cytoplasmic free-calcium, but ATP can increase the expressions of these proteins. PM2.5 exposure increased the levels of Th2-related cytokines of bronchoalveolar lavage fluid, lung inflammation, collagen deposition and hyperplasisa of goblet cells. Interestingly, the administration of ATP showed an inhibitory effect on lung inflammation induced by PM2.5. Together, our study reveals that PM2.5 impairs the ATP-induced transepithelial anion Isc through downregulating P2Y2R/CFTR pathway, and this process may participate in aggravating airway hyperresponsiveness and airway inflammation. These findings may provide important insights on PM2.5-mediated airway epithelial injury.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
International Journal of Medical Sciences
International Journal of Medical Sciences MEDICINE, GENERAL & INTERNAL-
CiteScore
7.20
自引率
0.00%
发文量
185
审稿时长
2.7 months
期刊介绍: Original research papers, reviews, and short research communications in any medical related area can be submitted to the Journal on the understanding that the work has not been published previously in whole or part and is not under consideration for publication elsewhere. Manuscripts in basic science and clinical medicine are both considered. There is no restriction on the length of research papers and reviews, although authors are encouraged to be concise. Short research communication is limited to be under 2500 words.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信