N6-甲基腺苷修饰的SRPK1通过PKM剪接促进肺腺癌的有氧糖酵解

IF 9.2 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY
Anqi Wang, Yuanyuan Zeng, Weijie Zhang, Jian Zhao, Lirong Gao, Jianjun Li, Jianjie Zhu, Zeyi Liu, Jian-An Huang
{"title":"N6-甲基腺苷修饰的SRPK1通过PKM剪接促进肺腺癌的有氧糖酵解","authors":"Anqi Wang, Yuanyuan Zeng, Weijie Zhang, Jian Zhao, Lirong Gao, Jianjun Li, Jianjie Zhu, Zeyi Liu, Jian-An Huang","doi":"10.1186/s11658-024-00622-5","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The RNA N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m<sup>6</sup>A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear.</p><p><strong>Methods: </strong>Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m<sup>6</sup>A epitranscriptomic microarray was utilized to the assess m<sup>6</sup>A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m<sup>6</sup>A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m<sup>6</sup>A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism.</p><p><strong>Results: </strong>The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m<sup>6</sup>A levels of SRPK1. Mechanistically, SRPK1's m<sup>6</sup>A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD.</p><p><strong>Conclusion: </strong>It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m<sup>6</sup>A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.</p>","PeriodicalId":9688,"journal":{"name":"Cellular & Molecular Biology Letters","volume":null,"pages":null},"PeriodicalIF":9.2000,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11295518/pdf/","citationCount":"0","resultStr":"{\"title\":\"N<sup>6</sup>-methyladenosine-modified SRPK1 promotes aerobic glycolysis of lung adenocarcinoma via PKM splicing.\",\"authors\":\"Anqi Wang, Yuanyuan Zeng, Weijie Zhang, Jian Zhao, Lirong Gao, Jianjun Li, Jianjie Zhu, Zeyi Liu, Jian-An Huang\",\"doi\":\"10.1186/s11658-024-00622-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>The RNA N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m<sup>6</sup>A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear.</p><p><strong>Methods: </strong>Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m<sup>6</sup>A epitranscriptomic microarray was utilized to the assess m<sup>6</sup>A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m<sup>6</sup>A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m<sup>6</sup>A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism.</p><p><strong>Results: </strong>The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m<sup>6</sup>A levels of SRPK1. Mechanistically, SRPK1's m<sup>6</sup>A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD.</p><p><strong>Conclusion: </strong>It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m<sup>6</sup>A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.</p>\",\"PeriodicalId\":9688,\"journal\":{\"name\":\"Cellular & Molecular Biology Letters\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":9.2000,\"publicationDate\":\"2024-08-02\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11295518/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cellular & Molecular Biology Letters\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1186/s11658-024-00622-5\",\"RegionNum\":1,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"BIOCHEMISTRY & MOLECULAR BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cellular & Molecular Biology Letters","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s11658-024-00622-5","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:RNA的N6-甲基腺苷(m6A)修饰已成为表观遗传学调控的一个重要热点。丝氨酸-精氨酸蛋白激酶 1(SRPK1)与多种癌症的发病机制有关。然而,SRPK1的m6A修饰及其与肺腺癌(LUAD)发病机制的关系仍不清楚:方法:采用 Western 印迹和聚合酶链反应(PCR)分析来确定基因和蛋白质的表达。进行了功能缺失和功能增益实验,以阐明 METTL3 和 SRPK1 对 LUAD 糖酵解和肿瘤发生的影响。通过RNA免疫沉淀(RIP)、m6A RNA免疫沉淀(MeRIP)和RNA稳定性测试,阐明了SRPK1介导的METTL3在LUAD中的m6A修饰机制。应用代谢定量和共免疫沉淀实验研究了SRPK1介导LUAD代谢的分子机制:结果:表转录组芯片分析表明,SRPK1在LUAD中可发生高甲基化和上调。主要的跨甲基化酶METTL3被上调,诱导了SRPK1的异常高m6A水平。从机制上讲,SRPK1的m6A位点直接被METTL3甲基化,METTL3还以一种依赖于IGF2BP2的方式稳定了SRPK1。甲基化的 SRPK1 随后通过增强糖酵解促进了 LUAD 的进展。进一步的代谢定量、共免疫沉淀和免疫印迹检测发现,SRPK1与PKM剪接的重要调节因子hnRNPA1相互作用,从而通过上调LUAD中的PKM2促进糖酵解。然而,METTL3抑制剂STM2457可以通过抑制LUAD中的SRPK1和糖酵解,在体外和体内逆转上述效应:结论:研究发现,在LUAD中,异常表达的METTL3通过m6A-IGF2BP2依赖性机制上调SRPK1水平。METTL3诱导的SRPK1通过增强糖酵解促进了LUAD细胞的增殖,METTL3的小分子抑制剂STM2457可能是治疗LUAD患者的另一种新策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
N6-methyladenosine-modified SRPK1 promotes aerobic glycolysis of lung adenocarcinoma via PKM splicing.

Background: The RNA N6-methyladenosine (m6A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m6A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear.

Methods: Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m6A epitranscriptomic microarray was utilized to the assess m6A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m6A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m6A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism.

Results: The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m6A levels of SRPK1. Mechanistically, SRPK1's m6A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD.

Conclusion: It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m6A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.

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
Cellular & Molecular Biology Letters
Cellular & Molecular Biology Letters 生物-生化与分子生物学
CiteScore
11.60
自引率
13.30%
发文量
101
审稿时长
3 months
期刊介绍: Cellular & Molecular Biology Letters is an international journal dedicated to the dissemination of fundamental knowledge in all areas of cellular and molecular biology, cancer cell biology, and certain aspects of biochemistry, biophysics and biotechnology.
×
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