Plac8-mediated autophagy regulates nasopharyngeal carcinoma cell function via AKT/mTOR pathway.

IF 5.3 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Journal of Cellular and Molecular Medicine Pub Date : 2020-07-01 Epub Date: 2020-05-29 DOI:10.1111/jcmm.15409
Mao-Ling Huang, Cheng-Lin Qi, You Zou, Rui Yang, Yang Jiang, Jian-Fei Sheng, Yong-Gang Kong, Ze-Zhang Tao, Shi-Ming Chen
{"title":"Plac8-mediated autophagy regulates nasopharyngeal carcinoma cell function via AKT/mTOR pathway.","authors":"Mao-Ling Huang,&nbsp;Cheng-Lin Qi,&nbsp;You Zou,&nbsp;Rui Yang,&nbsp;Yang Jiang,&nbsp;Jian-Fei Sheng,&nbsp;Yong-Gang Kong,&nbsp;Ze-Zhang Tao,&nbsp;Shi-Ming Chen","doi":"10.1111/jcmm.15409","DOIUrl":null,"url":null,"abstract":"<p><p>To explore the relationship between autophagy and cell function, we investigated how PLAC8-mediated autophagy influences proliferation, apoptosis and epithelial-mesenchymal transition (EMT) in NPC. Colony formation analyses and CCK8 assays were used to assess the proliferative capacity of NPC cells. Transmission electron microscopy (TEM) was used to identify autophagosomes. Autophagic flux was monitored using the tandem monomeric RFP-GFP-tagged LC3 (tfLC3) assay. The rate of apoptosis in NPC cells was analysed by flow cytometry. Western blot analysis was used to evaluate the activation of autophagy and the signalling status of the AKT/mTOR pathway. Our study reveals that knocking out PLAC8 (koPLAC8) induces autophagy and apoptosis, while suppressing NPC cell proliferation and EMT. However, inhibition of autophagy with 3-methyladenine or by knocking down Beclin-1 reverses the cell proliferation, apoptosis and EMT influenced by koPLAC8. We find that koPLAC8 inhibits the phosphorylation of AKT and its downstream target, mTOR. Moreover, immunofluorescence and co-immunoprecipitation reveal complete PLAC8/AKT colocalization and PLAC8/AKT interaction, respectively. Furthermore, knockout of PLAC8 induced autophagy and inactivated AKT/mTOR signalling pathway of NPC xenografts. Overall, our findings demonstrate that koPLAC8 induces autophagy via the AKT/mTOR pathway, thereby inhibiting cell proliferation and EMT, and promoting apoptosis in NPC cells.</p>","PeriodicalId":15215,"journal":{"name":"Journal of Cellular and Molecular Medicine","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2020-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/jcmm.15409","citationCount":"17","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Cellular and Molecular Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1111/jcmm.15409","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2020/5/29 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"Biochemistry, Genetics and Molecular Biology","Score":null,"Total":0}
引用次数: 17

Abstract

To explore the relationship between autophagy and cell function, we investigated how PLAC8-mediated autophagy influences proliferation, apoptosis and epithelial-mesenchymal transition (EMT) in NPC. Colony formation analyses and CCK8 assays were used to assess the proliferative capacity of NPC cells. Transmission electron microscopy (TEM) was used to identify autophagosomes. Autophagic flux was monitored using the tandem monomeric RFP-GFP-tagged LC3 (tfLC3) assay. The rate of apoptosis in NPC cells was analysed by flow cytometry. Western blot analysis was used to evaluate the activation of autophagy and the signalling status of the AKT/mTOR pathway. Our study reveals that knocking out PLAC8 (koPLAC8) induces autophagy and apoptosis, while suppressing NPC cell proliferation and EMT. However, inhibition of autophagy with 3-methyladenine or by knocking down Beclin-1 reverses the cell proliferation, apoptosis and EMT influenced by koPLAC8. We find that koPLAC8 inhibits the phosphorylation of AKT and its downstream target, mTOR. Moreover, immunofluorescence and co-immunoprecipitation reveal complete PLAC8/AKT colocalization and PLAC8/AKT interaction, respectively. Furthermore, knockout of PLAC8 induced autophagy and inactivated AKT/mTOR signalling pathway of NPC xenografts. Overall, our findings demonstrate that koPLAC8 induces autophagy via the AKT/mTOR pathway, thereby inhibiting cell proliferation and EMT, and promoting apoptosis in NPC cells.

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
plac8介导的自噬通过AKT/mTOR通路调控鼻咽癌细胞功能。
为了探讨自噬与细胞功能之间的关系,我们研究了plac8介导的自噬如何影响鼻咽癌细胞的增殖、凋亡和上皮间质转化(EMT)。使用集落形成分析和CCK8测定来评估鼻咽癌细胞的增殖能力。透射电镜(TEM)鉴定自噬体。自噬通量监测采用串联单体rfp - gfp标记LC3 (tfLC3)试验。流式细胞术分析鼻咽癌细胞凋亡率。Western blot分析自噬的激活情况及AKT/mTOR通路的信号传导状态。我们的研究表明,敲除PLAC8 (koPLAC8)可诱导自噬和凋亡,同时抑制鼻咽癌细胞的增殖和EMT。然而,用3-甲基腺嘌呤或敲低Beclin-1抑制自噬可以逆转koPLAC8对细胞增殖、凋亡和EMT的影响。我们发现koPLAC8抑制AKT及其下游靶点mTOR的磷酸化。此外,免疫荧光和共免疫沉淀分别显示了完整的PLAC8/AKT共定位和PLAC8/AKT相互作用。此外,敲除PLAC8可诱导鼻咽癌异种移植物的自噬和AKT/mTOR信号通路失活。总之,我们的研究结果表明,koPLAC8通过AKT/mTOR途径诱导自噬,从而抑制细胞增殖和EMT,促进鼻咽癌细胞凋亡。
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
CiteScore
10.00
自引率
1.90%
发文量
496
审稿时长
28 weeks
期刊介绍: Bridging physiology and cellular medicine, and molecular biology and molecular therapeutics, Journal of Cellular and Molecular Medicine publishes basic research that furthers our understanding of the cellular and molecular mechanisms of disease and translational studies that convert this knowledge into therapeutic approaches.
期刊最新文献
Downregulation of p300/CBP-associated factor inhibits cardiomyocyte apoptosis via suppression of NF-κB pathway in ischaemia/reperfusion injury rats. Application of Joint Mobilizing Chuna Following Tibial Plateau Fracture Surgery: A Study of Two Cases Korean Domestic Trends of Clinical Research and Direction of Intervention for Fibromyalgia Methylprednisolone alleviates multiple sclerosis by expanding myeloid-derived suppressor cells via glucocorticoid receptor β and S100A8/9 up-regulation. Loganetin and 5-fluorouracil synergistically inhibit the carcinogenesis of gastric cancer cells via down-regulation of the Wnt/β-catenin pathway.
×
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