Long non-coding RNA DNMBP-AS1 promotes prostate cancer development by regulating LCLAT1.

IF 2.1 4区 医学 Q3 ANDROLOGY
Xiangang Yin, Suying Wang, Rong Ge, Jinping Chen, Youliang Gao, Shanshan Xu, Ting Yang
{"title":"Long non-coding RNA DNMBP-AS1 promotes prostate cancer development by regulating LCLAT1.","authors":"Xiangang Yin,&nbsp;Suying Wang,&nbsp;Rong Ge,&nbsp;Jinping Chen,&nbsp;Youliang Gao,&nbsp;Shanshan Xu,&nbsp;Ting Yang","doi":"10.1080/19396368.2022.2129520","DOIUrl":null,"url":null,"abstract":"<p><p>Prostate cancer (PCa) is as a serious threat to male's health around the world. Recent studies have indicated that long non-coding RNAs (lncRNAs) occupy an important position in various human cancers. However, the function and mechanism of lncRNA DNMBP antisense RNA 1 (DNMBP-AS1) in PCa is rarely investigated. RT-qPCR analysis was used to test gene expression. CCK-8, colony formation, EdU staining and transwell assays were conducted to assess the function of DNMBP-AS1 on PCa cell behaviors. RNA pull down, RIP and luciferase reporter assays were implemented to verify the mechanism of DNMBP-AS1. DNMBP-AS1 was obviously up-regulated in PCa cell lines. Functionally, DNMBP-AS1 knockdown weakened cell proliferation, migration and invasion of PCa. Mechanistically, DNMBP-AS1 sponged microRNA-6766-3p (miR-6766-3p) to regulate lysocardiolipin acyltransferase 1 (LCLAT1) expression. Furthermore, DNMBP-AS1 could stabilize LCLAT1 expression by recruiting ELAV like RNA binding protein 1 (ELAVL1). Consequently, rescue assays demonstrated that DNMBP-AS1 regulated PCa cell proliferation, migration and invasion through enhancing LCLAT1 expression. Collectively, we elucidated the function and regulatory mechanism of DNMBP-AS1 and provided the first evidence of DNMBP-AS1 as a driver for PCa.</p>","PeriodicalId":22184,"journal":{"name":"Systems Biology in Reproductive Medicine","volume":null,"pages":null},"PeriodicalIF":2.1000,"publicationDate":"2023-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Systems Biology in Reproductive Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/19396368.2022.2129520","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/1/5 0:00:00","PubModel":"Epub","JCR":"Q3","JCRName":"ANDROLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Prostate cancer (PCa) is as a serious threat to male's health around the world. Recent studies have indicated that long non-coding RNAs (lncRNAs) occupy an important position in various human cancers. However, the function and mechanism of lncRNA DNMBP antisense RNA 1 (DNMBP-AS1) in PCa is rarely investigated. RT-qPCR analysis was used to test gene expression. CCK-8, colony formation, EdU staining and transwell assays were conducted to assess the function of DNMBP-AS1 on PCa cell behaviors. RNA pull down, RIP and luciferase reporter assays were implemented to verify the mechanism of DNMBP-AS1. DNMBP-AS1 was obviously up-regulated in PCa cell lines. Functionally, DNMBP-AS1 knockdown weakened cell proliferation, migration and invasion of PCa. Mechanistically, DNMBP-AS1 sponged microRNA-6766-3p (miR-6766-3p) to regulate lysocardiolipin acyltransferase 1 (LCLAT1) expression. Furthermore, DNMBP-AS1 could stabilize LCLAT1 expression by recruiting ELAV like RNA binding protein 1 (ELAVL1). Consequently, rescue assays demonstrated that DNMBP-AS1 regulated PCa cell proliferation, migration and invasion through enhancing LCLAT1 expression. Collectively, we elucidated the function and regulatory mechanism of DNMBP-AS1 and provided the first evidence of DNMBP-AS1 as a driver for PCa.

长非编码RNA DNMBP-AS1通过调节LCLAT1促进前列腺癌症的发展。
癌症(PCa)是世界各地男性健康的严重威胁。最近的研究表明,长非编码RNA(lncRNA)在各种人类癌症中占有重要地位。然而,lncRNA-DNMBP反义RNA1(DNMBP-AS1)在前列腺癌中的作用和机制很少被研究。RT-qPCR分析用于检测基因表达。进行CCK-8、集落形成、EdU染色和transwell分析以评估DNMBP-AS1对PCa细胞行为的功能。进行RNA下拉、RIP和荧光素酶报告基因测定以验证DNMBP-AS1的机制。DNMBP-AS1在前列腺癌细胞系中明显上调。从功能上讲,DNMBP-AS1敲低削弱了PCa的细胞增殖、迁移和侵袭。从机制上讲,DNMBP-AS1吸收微小RNA-6766-3p(miR-6766-3ps)来调节赖氨酸甘油酰基转移酶1(LCLAT1)的表达。此外,DNMBP-AS1可以通过募集ELAV-like RNA结合蛋白1(ELAVL1)来稳定LCLAT1的表达。因此,拯救试验表明,DNMBP-AS1通过增强LCLAT1的表达来调节PCa细胞的增殖、迁移和侵袭。总之,我们阐明了DNMBP-AS1的功能和调节机制,并提供了DNMBP-AS1作为PCa驱动因素的第一个证据。
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 求助全文
来源期刊
CiteScore
4.30
自引率
4.20%
发文量
27
审稿时长
>12 weeks
期刊介绍: Systems Biology in Reproductive Medicine, SBiRM, publishes Research Articles, Communications, Applications Notes that include protocols a Clinical Corner that includes case reports, Review Articles and Hypotheses and Letters to the Editor on human and animal reproduction. The journal will highlight the use of systems approaches including genomic, cellular, proteomic, metabolomic, bioinformatic, molecular, and biochemical, to address fundamental questions in reproductive biology, reproductive medicine, and translational research. The journal publishes research involving human and animal gametes, stem cells, developmental biology and toxicology, and clinical care in reproductive medicine. Specific areas of interest to the journal include: male factor infertility and germ cell biology, reproductive technologies (gamete micro-manipulation and cryopreservation, in vitro fertilization/embryo transfer (IVF/ET) and contraception. Research that is directed towards developing new or enhanced technologies for clinical medicine or scientific research in reproduction is of significant interest to the journal.
×
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