三碘甲状腺原氨酸作用于 DAO,通过 p53/p21 信号通路促进细胞衰老,从而调控肺纤维化进程

IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Frontiers in Pharmacology Pub Date : 2024-09-11 DOI:10.3389/fphar.2024.1433186
Xiaoshu Guo, Kai Xu, Lan Wang, Linke Ding, Wenwen Li, Xinsheng Zhang, Weiming Zhao, Ningdan Wang, Gaiping Wang, Wenyu Zhao, Ivan Rosas, Guoying Yu
{"title":"三碘甲状腺原氨酸作用于 DAO,通过 p53/p21 信号通路促进细胞衰老,从而调控肺纤维化进程","authors":"Xiaoshu Guo, Kai Xu, Lan Wang, Linke Ding, Wenwen Li, Xinsheng Zhang, Weiming Zhao, Ningdan Wang, Gaiping Wang, Wenyu Zhao, Ivan Rosas, Guoying Yu","doi":"10.3389/fphar.2024.1433186","DOIUrl":null,"url":null,"abstract":"BackgroundIdiopathic pulmonary fibrosis (IPF) is the result of multiple cycles of epithelial cell injury and fibroblast activation; currently, there is no clear etiology. Increasing evidence suggests that protein metabolism and amino acids play a crucial role in IPF, but the role of D-amino acids is not yet clear. The aim of this study was to identify novel mediators in order to test the hypothesis that D-amino acid oxidase (DAO) plays a significant role in the pathogenesis of IPF.MethodsWe analyzed DAO gene expression in patients with IPF and mice with bleomycin (BLM)-induced lung fibrosis. We performed <jats:italic>in vitro</jats:italic> and <jats:italic>in vivo</jats:italic> assays to determine the effect of DAO on primary type II alveolar epithelial cells from mice and A549 cells.ResultsDAO expression was downregulated in the lungs of IPF patients and BLM-induced fibrotic mice. Treatment with D-serine (D-Ser) or drug inhibition of DAO promoted cell senescence through the p53/p21 pathway. <jats:italic>Dao</jats:italic><jats:sup>−/−</jats:sup> mice showed an intensified fibrotic response, and the anti-fibrotic role of T<jats:sub>3</jats:sub> was abolished.ConclusionWe concluded that the DAO-p53/p21 axis might be a key anti-fibrotic pathway regulating the progress of fibrosis and facilitating the therapeutic role of T<jats:sub>3</jats:sub>.","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":null,"pages":null},"PeriodicalIF":4.4000,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Triiodothyronine acts on DAO to regulate pulmonary fibrosis progression by facilitating cell senescence through the p53/p21 signaling pathway\",\"authors\":\"Xiaoshu Guo, Kai Xu, Lan Wang, Linke Ding, Wenwen Li, Xinsheng Zhang, Weiming Zhao, Ningdan Wang, Gaiping Wang, Wenyu Zhao, Ivan Rosas, Guoying Yu\",\"doi\":\"10.3389/fphar.2024.1433186\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"BackgroundIdiopathic pulmonary fibrosis (IPF) is the result of multiple cycles of epithelial cell injury and fibroblast activation; currently, there is no clear etiology. Increasing evidence suggests that protein metabolism and amino acids play a crucial role in IPF, but the role of D-amino acids is not yet clear. The aim of this study was to identify novel mediators in order to test the hypothesis that D-amino acid oxidase (DAO) plays a significant role in the pathogenesis of IPF.MethodsWe analyzed DAO gene expression in patients with IPF and mice with bleomycin (BLM)-induced lung fibrosis. We performed <jats:italic>in vitro</jats:italic> and <jats:italic>in vivo</jats:italic> assays to determine the effect of DAO on primary type II alveolar epithelial cells from mice and A549 cells.ResultsDAO expression was downregulated in the lungs of IPF patients and BLM-induced fibrotic mice. Treatment with D-serine (D-Ser) or drug inhibition of DAO promoted cell senescence through the p53/p21 pathway. <jats:italic>Dao</jats:italic><jats:sup>−/−</jats:sup> mice showed an intensified fibrotic response, and the anti-fibrotic role of T<jats:sub>3</jats:sub> was abolished.ConclusionWe concluded that the DAO-p53/p21 axis might be a key anti-fibrotic pathway regulating the progress of fibrosis and facilitating the therapeutic role of T<jats:sub>3</jats:sub>.\",\"PeriodicalId\":12491,\"journal\":{\"name\":\"Frontiers in Pharmacology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":4.4000,\"publicationDate\":\"2024-09-11\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Frontiers in Pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.3389/fphar.2024.1433186\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in Pharmacology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.3389/fphar.2024.1433186","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

背景特发性肺纤维化(IPF)是上皮细胞损伤和成纤维细胞活化多重循环的结果;目前尚无明确的病因。越来越多的证据表明,蛋白质代谢和氨基酸在 IPF 中起着至关重要的作用,但 D-氨基酸的作用尚不明确。本研究旨在鉴定新的介质,以验证 D-氨基酸氧化酶(DAO)在 IPF 发病机制中发挥重要作用的假设。结果DAO在IPF患者和博莱霉素诱导的肺纤维化小鼠的肺中表达下调。用D-丝氨酸(D-Ser)处理或药物抑制DAO可通过p53/p21途径促进细胞衰老。结论我们认为,DAO-p53/p21轴可能是调节纤维化进程和促进T3治疗作用的关键抗纤维化途径。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Triiodothyronine acts on DAO to regulate pulmonary fibrosis progression by facilitating cell senescence through the p53/p21 signaling pathway
BackgroundIdiopathic pulmonary fibrosis (IPF) is the result of multiple cycles of epithelial cell injury and fibroblast activation; currently, there is no clear etiology. Increasing evidence suggests that protein metabolism and amino acids play a crucial role in IPF, but the role of D-amino acids is not yet clear. The aim of this study was to identify novel mediators in order to test the hypothesis that D-amino acid oxidase (DAO) plays a significant role in the pathogenesis of IPF.MethodsWe analyzed DAO gene expression in patients with IPF and mice with bleomycin (BLM)-induced lung fibrosis. We performed in vitro and in vivo assays to determine the effect of DAO on primary type II alveolar epithelial cells from mice and A549 cells.ResultsDAO expression was downregulated in the lungs of IPF patients and BLM-induced fibrotic mice. Treatment with D-serine (D-Ser) or drug inhibition of DAO promoted cell senescence through the p53/p21 pathway. Dao−/− mice showed an intensified fibrotic response, and the anti-fibrotic role of T3 was abolished.ConclusionWe concluded that the DAO-p53/p21 axis might be a key anti-fibrotic pathway regulating the progress of fibrosis and facilitating the therapeutic role of T3.
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
Frontiers in Pharmacology
Frontiers in Pharmacology PHARMACOLOGY & PHARMACY-
CiteScore
7.80
自引率
8.90%
发文量
5163
审稿时长
14 weeks
期刊介绍: Frontiers in Pharmacology is a leading journal in its field, publishing rigorously peer-reviewed research across disciplines, including basic and clinical pharmacology, medicinal chemistry, pharmacy and toxicology. Field Chief Editor Heike Wulff at UC Davis is supported by an outstanding Editorial Board of international researchers. This multidisciplinary open-access journal is at the forefront of disseminating and communicating scientific knowledge and impactful discoveries to researchers, academics, clinicians and the public worldwide.
期刊最新文献
The glucocorticoid dose-mortality nexus in pneumonia patients: unveiling the threshold effect. The impact of EU public procurement regulations on tenders in Spain: a study with adalimumab. The role of TRPV1 in chronic prostatitis: a review. Transcriptomics-based anti-tuberculous mechanism of traditional Chinese polyherbal preparation NiuBeiXiaoHe intermediates. Applications and challenges of photodynamic therapy in the treatment of skin malignancies.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1