METTL3/MALAT1/ELAVL1轴促进卵巢癌的肿瘤生长

IF 2.7 4区 医学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY
OncoTargets and therapy Pub Date : 2024-02-08 eCollection Date: 2024-01-01 DOI:10.2147/OTT.S431810
Jian Xiong, Wenqin Lian, Rui Zhao, Kefei Gao
{"title":"METTL3/MALAT1/ELAVL1轴促进卵巢癌的肿瘤生长","authors":"Jian Xiong, Wenqin Lian, Rui Zhao, Kefei Gao","doi":"10.2147/OTT.S431810","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Studies increasingly recognize the role of N6-methyladenosine (<i>m6A</i>) modification in cancer occurrence and development. METTL3 is a core catalytic subunit of m6A-modified methyltransferases complex, but its regulatory mechanism in ovarian cancer (OC) is not clear.</p><p><strong>Methods: </strong>In this study, GEPIA 2.0 database was applied for expression analysis, survival analysis and correlation analysis for OC. Additionally, in vitro and in vivo assays were conducted to explore regulatory mechanisms of METTL3 in OC.</p><p><strong>Results: </strong>We found that METTL3 and <i>MALAT1</i> were significantly overexpressed in OC tissues and cells compared to normal ovarian tissues and cells. The proliferation rate of OC cells was reduced significantly after knocking down the expression of <i>METTL3</i> or <i>MALAT1</i>. Subsequently, <i>MALAT1</i> as oncogene was found to interact with METTL3 and was upregulated in OC tissues and cells. Silencing <i>MALAT1</i> inhibited OC cell proliferation. Further studies indicated that METTL3 enhanced the stability of <i>MALAT1</i> by promoting the m6A modification of <i>MALAT1</i> and that ELAVL1 as a downstream binding protein significantly up-regulated <i>MALAT1</i> expression.</p><p><strong>Conclusion: </strong>In conclusion, METTL3 was a carcinogenic molecule that promoted the occurrence of OC. The potential mechanism of the carcinogenic effect of METTL3 was realized by enhancing the m6A modification of <i>MALAT1</i> mRNA through RNA binding protein ELAVL1.</p>","PeriodicalId":19534,"journal":{"name":"OncoTargets and therapy","volume":null,"pages":null},"PeriodicalIF":2.7000,"publicationDate":"2024-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10860502/pdf/","citationCount":"0","resultStr":"{\"title\":\"METTL3/<i>MALAT1</i>/ELAVL1 Axis Promotes Tumor Growth in Ovarian Cancer.\",\"authors\":\"Jian Xiong, Wenqin Lian, Rui Zhao, Kefei Gao\",\"doi\":\"10.2147/OTT.S431810\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Studies increasingly recognize the role of N6-methyladenosine (<i>m6A</i>) modification in cancer occurrence and development. METTL3 is a core catalytic subunit of m6A-modified methyltransferases complex, but its regulatory mechanism in ovarian cancer (OC) is not clear.</p><p><strong>Methods: </strong>In this study, GEPIA 2.0 database was applied for expression analysis, survival analysis and correlation analysis for OC. Additionally, in vitro and in vivo assays were conducted to explore regulatory mechanisms of METTL3 in OC.</p><p><strong>Results: </strong>We found that METTL3 and <i>MALAT1</i> were significantly overexpressed in OC tissues and cells compared to normal ovarian tissues and cells. The proliferation rate of OC cells was reduced significantly after knocking down the expression of <i>METTL3</i> or <i>MALAT1</i>. Subsequently, <i>MALAT1</i> as oncogene was found to interact with METTL3 and was upregulated in OC tissues and cells. Silencing <i>MALAT1</i> inhibited OC cell proliferation. Further studies indicated that METTL3 enhanced the stability of <i>MALAT1</i> by promoting the m6A modification of <i>MALAT1</i> and that ELAVL1 as a downstream binding protein significantly up-regulated <i>MALAT1</i> expression.</p><p><strong>Conclusion: </strong>In conclusion, METTL3 was a carcinogenic molecule that promoted the occurrence of OC. The potential mechanism of the carcinogenic effect of METTL3 was realized by enhancing the m6A modification of <i>MALAT1</i> mRNA through RNA binding protein ELAVL1.</p>\",\"PeriodicalId\":19534,\"journal\":{\"name\":\"OncoTargets and therapy\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.7000,\"publicationDate\":\"2024-02-08\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10860502/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"OncoTargets and therapy\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.2147/OTT.S431810\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q3\",\"JCRName\":\"BIOTECHNOLOGY & APPLIED MICROBIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"OncoTargets and therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.2147/OTT.S431810","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q3","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:越来越多的研究认识到N6-甲基腺苷(m6A)修饰在癌症发生和发展中的作用。METTL3是m6A修饰甲基转移酶复合物的核心催化亚基,但其在卵巢癌(OC)中的调控机制尚不清楚:本研究应用 GEPIA 2.0 数据库对 OC 进行了表达分析、生存分析和相关分析。结果:我们发现,METTL3与卵巢癌的相关性很高,而METTL3与卵巢癌的相关性很低:结果:我们发现,与正常卵巢组织和细胞相比,METTL3和MALAT1在OC组织和细胞中明显高表达。结果:我们发现,与正常卵巢组织和细胞相比,METTL3 和 MALAT1 在 OC 组织和细胞中明显高表达。随后发现,MALAT1作为癌基因与METTL3相互作用,并在OC组织和细胞中上调。抑制 MALAT1 可抑制 OC 细胞增殖。进一步研究表明,METTL3通过促进MALAT1的m6A修饰增强了MALAT1的稳定性,而ELAVL1作为下游结合蛋白可显著上调MALAT1的表达:总之,METTL3是一种促进OC发生的致癌分子。METTL3致癌作用的潜在机制是通过RNA结合蛋白ELAVL1增强MALAT1 mRNA的m6A修饰。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
METTL3/MALAT1/ELAVL1 Axis Promotes Tumor Growth in Ovarian Cancer.

Background: Studies increasingly recognize the role of N6-methyladenosine (m6A) modification in cancer occurrence and development. METTL3 is a core catalytic subunit of m6A-modified methyltransferases complex, but its regulatory mechanism in ovarian cancer (OC) is not clear.

Methods: In this study, GEPIA 2.0 database was applied for expression analysis, survival analysis and correlation analysis for OC. Additionally, in vitro and in vivo assays were conducted to explore regulatory mechanisms of METTL3 in OC.

Results: We found that METTL3 and MALAT1 were significantly overexpressed in OC tissues and cells compared to normal ovarian tissues and cells. The proliferation rate of OC cells was reduced significantly after knocking down the expression of METTL3 or MALAT1. Subsequently, MALAT1 as oncogene was found to interact with METTL3 and was upregulated in OC tissues and cells. Silencing MALAT1 inhibited OC cell proliferation. Further studies indicated that METTL3 enhanced the stability of MALAT1 by promoting the m6A modification of MALAT1 and that ELAVL1 as a downstream binding protein significantly up-regulated MALAT1 expression.

Conclusion: In conclusion, METTL3 was a carcinogenic molecule that promoted the occurrence of OC. The potential mechanism of the carcinogenic effect of METTL3 was realized by enhancing the m6A modification of MALAT1 mRNA through RNA binding protein ELAVL1.

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
OncoTargets and therapy
OncoTargets and therapy BIOTECHNOLOGY & APPLIED MICROBIOLOGY-ONCOLOGY
CiteScore
9.70
自引率
0.00%
发文量
221
审稿时长
1 months
期刊介绍: OncoTargets and Therapy is an international, peer-reviewed journal focusing on molecular aspects of cancer research, that is, the molecular diagnosis of and targeted molecular or precision therapy for all types of cancer. The journal is characterized by the rapid reporting of high-quality original research, basic science, reviews and evaluations, expert opinion and commentary that shed novel insight on a cancer or cancer subtype. Specific topics covered by the journal include: -Novel therapeutic targets and innovative agents -Novel therapeutic regimens for improved benefit and/or decreased side effects -Early stage clinical trials Further considerations when submitting to OncoTargets and Therapy: -Studies containing in vivo animal model data will be considered favorably. -Tissue microarray analyses will not be considered except in cases where they are supported by comprehensive biological studies involving multiple cell lines. -Biomarker association studies will be considered only when validated by comprehensive in vitro data and analysis of human tissue samples. -Studies utilizing publicly available data (e.g. GWAS/TCGA/GEO etc.) should add to the body of knowledge about a specific disease or relevant phenotype and must be validated using the authors’ own data through replication in an independent sample set and functional follow-up. -Bioinformatics studies must be validated using the authors’ own data through replication in an independent sample set and functional follow-up. -Single nucleotide polymorphism (SNP) studies will not be considered.
×
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