Deubiquitinase USP18 mediates cell migration, apoptosis and ferroptosis in lung adenocarcinoma by depending on POU4F1/PRKAA2 axis.

IF 3.4 2区 医学 Q2 ONCOLOGY BMC Cancer Pub Date : 2025-03-23 DOI:10.1186/s12885-025-13869-8
Xinping Pan, Hui Deng
{"title":"Deubiquitinase USP18 mediates cell migration, apoptosis and ferroptosis in lung adenocarcinoma by depending on POU4F1/PRKAA2 axis.","authors":"Xinping Pan, Hui Deng","doi":"10.1186/s12885-025-13869-8","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Lung adenocarcinoma (LUAD) is a common type of lung cancer and its pathogenic mechanism is complicated. A profound research for the molecular mechanism in LUAD is indispensable.</p><p><strong>Methods: </strong>Gene levels were detected via real-time quantitative polymerase chain reaction and western blot. Proliferation, migration and apoptosis were assessed using colony formation assay, wound healing assay, and flow cytometry. Ferroptosis was evaluated through oxidative stress and iron level. Relations between genes were analyzed using Immunoprecipitation (IP) assay and ubiquitination assay, as well as ChIP assay and dual-luciferase reporter assay. USP18 function in vivo was explored using xenograft model.</p><p><strong>Results: </strong>Ubiquitin-specific protease 18 (USP18) was overexpressed in LUAD tissues and cells. LUAD cell proliferation and migration were suppressed but apoptosis and ferroptosis were enhanced after USP18 knockdown. Pou domain, class 4, transcription factor 1 (POU4F1) protein expression was stabilized through USP18-mediated deubiquitination. Function of USP18 silence was reversed by POU4F1 overexpression in LUAD cells. POU4F1 promoted transcription of AMPK-α2 (PRKAA2) and USP18 modulated PRKAA2 protein level via affecting POU4F1. POU4F1 regulated LUAD cell behaviors by upregulating PRKAA2. USP18 enhanced tumor growth in vivo via mediating POU4F1 and PRKAA2.</p><p><strong>Conclusion: </strong>All data demonstrated that USP18 acted as an oncogene in LUAD via interacting with POU4F1/PRKAA2 axis.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":"25 1","pages":"528"},"PeriodicalIF":3.4000,"publicationDate":"2025-03-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11929989/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"BMC Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12885-025-13869-8","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Lung adenocarcinoma (LUAD) is a common type of lung cancer and its pathogenic mechanism is complicated. A profound research for the molecular mechanism in LUAD is indispensable.

Methods: Gene levels were detected via real-time quantitative polymerase chain reaction and western blot. Proliferation, migration and apoptosis were assessed using colony formation assay, wound healing assay, and flow cytometry. Ferroptosis was evaluated through oxidative stress and iron level. Relations between genes were analyzed using Immunoprecipitation (IP) assay and ubiquitination assay, as well as ChIP assay and dual-luciferase reporter assay. USP18 function in vivo was explored using xenograft model.

Results: Ubiquitin-specific protease 18 (USP18) was overexpressed in LUAD tissues and cells. LUAD cell proliferation and migration were suppressed but apoptosis and ferroptosis were enhanced after USP18 knockdown. Pou domain, class 4, transcription factor 1 (POU4F1) protein expression was stabilized through USP18-mediated deubiquitination. Function of USP18 silence was reversed by POU4F1 overexpression in LUAD cells. POU4F1 promoted transcription of AMPK-α2 (PRKAA2) and USP18 modulated PRKAA2 protein level via affecting POU4F1. POU4F1 regulated LUAD cell behaviors by upregulating PRKAA2. USP18 enhanced tumor growth in vivo via mediating POU4F1 and PRKAA2.

Conclusion: All data demonstrated that USP18 acted as an oncogene in LUAD via interacting with POU4F1/PRKAA2 axis.

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
去泛素酶USP18依赖于POU4F1/PRKAA2轴介导肺腺癌细胞迁移、凋亡和铁凋亡。
背景:肺腺癌(LUAD)是一种常见的肺癌类型,其发病机制复杂。深入研究LUAD的分子机制是必不可少的。方法:采用实时定量聚合酶链反应和western blot检测基因水平。使用菌落形成实验、伤口愈合实验和流式细胞术评估增殖、迁移和凋亡。通过氧化应激和铁水平评价铁下垂。采用免疫沉淀(IP)法、泛素化法、ChIP法和双荧光素酶报告基因法分析基因间的关系。采用异种移植模型探讨USP18在体内的功能。结果:泛素特异性蛋白酶18 (USP18)在LUAD组织和细胞中过表达。敲除USP18后,LUAD细胞增殖和迁移受到抑制,但凋亡和铁下垂增强。通过usp18介导的去泛素化,稳定了4类Pou域转录因子1 (POU4F1)蛋白的表达。在LUAD细胞中,POU4F1过表达可逆转USP18沉默的功能。POU4F1促进AMPK-α2 (PRKAA2)转录,USP18通过影响POU4F1调节PRKAA2蛋白水平。POU4F1通过上调PRKAA2调控LUAD细胞行为。USP18通过介导POU4F1和PRKAA2在体内促进肿瘤生长。结论:所有数据均表明USP18通过与POU4F1/PRKAA2轴相互作用在LUAD中发挥致癌基因的作用。
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
BMC Cancer
BMC Cancer 医学-肿瘤学
CiteScore
6.00
自引率
2.60%
发文量
1204
审稿时长
6.8 months
期刊介绍: BMC Cancer is an open access, peer-reviewed journal that considers articles on all aspects of cancer research, including the pathophysiology, prevention, diagnosis and treatment of cancers. The journal welcomes submissions concerning molecular and cellular biology, genetics, epidemiology, and clinical trials.
期刊最新文献
Investigating the causal link between metformin and lung cancer risk: a two-sample mendelian randomization analysis. Incidence, patterns of clinical presentation, and haematological characteristics of paediatric acute myeloid leukaemia in Uganda: a retrospective analysis. Supervised machine learning to quantitatively assess the prognostic value of tumor-infiltrating lymphocytes in gastric cancer. Interpretable machine learning using CT radiomics predicts pathological upgrading after secondary resection in non-muscle-invasive bladder cancer. Efficacy and safety of 7-day aerosolized epigallocatechin-3-gallate in oncologic patients with COVID-19 pneumonia.
×
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:604180095
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