Biological Evaluation of Alkyl Triphenylphosphonium Ostruthin Derivatives as Potential Anti-Inflammatory Agents Targeting the Nuclear Factor κB Signaling Pathway in Human Lung Adenocarcinoma A549 Cells

BioChem Pub Date : 2021-01-01 DOI:10.3390/biochem1020010
Nghia T. Vo, Eiichi Kusagawa, Kaori Nakano, Chihiro Moriwaki, Yasunobu Miyake, Sayaka Haruyama, Sayuri Fukuhara, N. Nguyen, P. Dang, M. T. T. Nguyen, T. Kataoka
{"title":"Biological Evaluation of Alkyl Triphenylphosphonium Ostruthin Derivatives as Potential Anti-Inflammatory Agents Targeting the Nuclear Factor κB Signaling Pathway in Human Lung Adenocarcinoma A549 Cells","authors":"Nghia T. Vo, Eiichi Kusagawa, Kaori Nakano, Chihiro Moriwaki, Yasunobu Miyake, Sayaka Haruyama, Sayuri Fukuhara, N. Nguyen, P. Dang, M. T. T. Nguyen, T. Kataoka","doi":"10.3390/biochem1020010","DOIUrl":null,"url":null,"abstract":"Ostruthin (6-geranyl-7-hydroxycoumarin) is one of the constituents isolated from Paramignya trimera and has been classified as a simple coumarin. We recently reported the synthesis of alkyl triphenylphosphonium (TPP) derivatives from ostruthin and evaluated their anticancer activities. In the present study, we demonstrated that alkyl TPP ostruthin derivatives inhibited the up-regulation of cell-surface intercellular adhesion molecule-1 (ICAM-1) in human lung adenocarcinoma A549 cells stimulated with tumor necrosis factor-α (TNF-α) without affecting cell viability, while ostruthin itself exerted cytotoxicity against A549 cells. The heptyl TPP ostruthin derivative (termed OS8) attenuated the up-regulation of ICAM-1 mRNA expression at concentrations higher than 40 µM in TNF-α-stimulated A549 cells. OS8 inhibited TNF-α-induced nuclear factor κB (NF-κB)-responsive luciferase reporter activity at concentrations higher than 40 µM, but did not affect the translocation of the NF-κB subunit RelA in response to the TNF-α stimulation at concentrations up to 100 µM. A chromatin immunoprecipitation assay showed that OS8 at 100 µM prevented the binding of RelA to the ICAM-1 promoter. We also showed that OS8 at 100 µM inhibited the TNF-α-induced phosphorylation of RelA at Ser 536. Moreover, the TNF-α-induced phosphorylation of an inhibitor of NF-κB α and extracellular signal-regulated kinase was reduced by OS8. These results indicate that OS8 has potential as an anti-inflammatory agent that targets the NF-κB signaling pathway.","PeriodicalId":72357,"journal":{"name":"BioChem","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"1","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"BioChem","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3390/biochem1020010","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 1

Abstract

Ostruthin (6-geranyl-7-hydroxycoumarin) is one of the constituents isolated from Paramignya trimera and has been classified as a simple coumarin. We recently reported the synthesis of alkyl triphenylphosphonium (TPP) derivatives from ostruthin and evaluated their anticancer activities. In the present study, we demonstrated that alkyl TPP ostruthin derivatives inhibited the up-regulation of cell-surface intercellular adhesion molecule-1 (ICAM-1) in human lung adenocarcinoma A549 cells stimulated with tumor necrosis factor-α (TNF-α) without affecting cell viability, while ostruthin itself exerted cytotoxicity against A549 cells. The heptyl TPP ostruthin derivative (termed OS8) attenuated the up-regulation of ICAM-1 mRNA expression at concentrations higher than 40 µM in TNF-α-stimulated A549 cells. OS8 inhibited TNF-α-induced nuclear factor κB (NF-κB)-responsive luciferase reporter activity at concentrations higher than 40 µM, but did not affect the translocation of the NF-κB subunit RelA in response to the TNF-α stimulation at concentrations up to 100 µM. A chromatin immunoprecipitation assay showed that OS8 at 100 µM prevented the binding of RelA to the ICAM-1 promoter. We also showed that OS8 at 100 µM inhibited the TNF-α-induced phosphorylation of RelA at Ser 536. Moreover, the TNF-α-induced phosphorylation of an inhibitor of NF-κB α and extracellular signal-regulated kinase was reduced by OS8. These results indicate that OS8 has potential as an anti-inflammatory agent that targets the NF-κB signaling pathway.
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
烷基三苯基磷ostrutin衍生物作为靶向人肺腺癌A549细胞核因子κB信号通路的潜在抗炎药的生物学评价
Ostruthin (6-geranyl-7-hydroxycoumarin)是从Paramignya trimera中分离得到的一种简单的香豆素。本文报道了以鱼胆素为原料合成烷基三苯磷(TPP)衍生物,并对其抗癌活性进行了评价。在本研究中,我们证明烷基TPP ostrutin衍生物抑制肿瘤坏死因子-α (TNF-α)刺激的人肺腺癌A549细胞中细胞表面细胞间粘附分子-1 (ICAM-1)的上调而不影响细胞活力,而ostrutin本身对A549细胞具有细胞毒性。在TNF-α-刺激的A549细胞中,庚基TPP蛋白衍生物(称为OS8)在浓度高于40µM时减弱了ICAM-1 mRNA表达的上调。OS8在浓度高于40µM时抑制TNF-α-诱导的核因子κB (NF-κB)反应性荧光素酶报告活性,但在浓度高达100µM时不影响NF-κB亚基RelA对TNF-α刺激的易位。染色质免疫沉淀实验表明,OS8在100µM时阻止RelA与ICAM-1启动子结合。我们还发现,100µM的OS8可以抑制TNF-α-诱导的RelA丝氨酸536位点的磷酸化。此外,TNF-α-诱导的NF-κB α和细胞外信号调节激酶抑制剂的磷酸化被OS8降低。这些结果表明OS8可能作为一种靶向NF-κB信号通路的抗炎药。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Evaluation of Antioxidant, Antibacterial and Enzyme-Inhibitory Properties of Dittany and Thyme Extracts and Their Application in Hydrogel Preparation Sphingolipid Signaling and Complement Activation in Glioblastoma: A Promising Avenue for Therapeutic Intervention Novel Tetrazolium-Based Colorimetric Assay for Helicase nsp13 in SARS-CoV-2 Bioinformatic Analysis of Metabolomic Data: From Raw Spectra to Biological Insight New Insights into Hsp90 Structural Plasticity Revealed by cryoEM
×
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