Targeting N4-acetylcytidine suppresses hepatocellular carcinoma progression by repressing eEF2-mediated HMGB2 mRNA translation.

IF 20.1 1区 医学 Q1 ONCOLOGY
Hailing Liu, Lei Xu, Shiwei Yue, Hongfei Su, Xing Chen, Qiumeng Liu, Hui Li, Huifang Liang, Xiaoping Chen, Jiefeng He, Zeyang Ding, Bixiang Zhang
{"title":"Targeting N4-acetylcytidine suppresses hepatocellular carcinoma progression by repressing eEF2-mediated HMGB2 mRNA translation.","authors":"Hailing Liu, Lei Xu, Shiwei Yue, Hongfei Su, Xing Chen, Qiumeng Liu, Hui Li, Huifang Liang, Xiaoping Chen, Jiefeng He, Zeyang Ding, Bixiang Zhang","doi":"10.1002/cac2.12595","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>N4-acetylcytidine (ac4C) represents a novel messenger RNA (mRNA) modification, and its associated acetyltransferase N-acetyltransferase 10 (NAT10) plays a crucial role in the initiation and progression of tumors by regulating mRNA functionality. However, its role in hepatocellular carcinoma (HCC) development and prognosis is largely unknown. This study aimed to elucidate the role of NAT10-mediated ac4C in HCC progression and provide a promising therapeutic approach.</p><p><strong>Methods: </strong>The ac4C levels were evaluated by dot blot and ultra-performance liquid chromatography-tandem mass spectrometry with harvested HCC tissues. The expression of NAT10 was investigated using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemical staining across 91 cohorts of HCC patients. To explore the underlying mechanisms of NAT10-ac4C in HCC, we employed a comprehensive approach integrating acetylated RNA immunoprecipitation and sequencing, RNA sequencing and ribosome profiling analyses, along with RNA immunoprecipitation, RNA pull-down, mass spectrometry, and site-specific mutation analyses. The drug affinity responsive targets stability, cellular thermal shift assay, and surface plasmon resonance assays were performed to assess the specific binding of NAT10 and Panobinostat. Furthermore, the efficacy of targeting NAT10-ac4C for HCC treatment was elucidated through in vitro experiments using HCC cells and in vivo HCC mouse models.</p><p><strong>Results: </strong>Our investigation revealed a significant increase in both the ac4C RNA level and NAT10 expression in HCC. Notably, elevated NAT10 expression was associated with poor outcomes in HCC patients. Functionally, silencing NAT10 suppressed HCC proliferation and metastasis in vitro and in vivo. Mechanistically, NAT10 stimulates the ac4C modification within the coding sequence (CDS) of high mobility group protein B2 (HMGB2), which subsequently enhances HMGB2 translation by facilitating eukaryotic elongation factor 2 (eEF2) binding to the ac4C sites on HMGB2 mRNA's CDS. Additionally, high-throughput compound library screening revealed Panobinostat as a potent inhibitor of NAT10-mediated ac4C modification. This inhibition significantly attenuated HCC growth and metastasis in both in vitro experiments using HCC cells and in vivo HCC mouse models.</p><p><strong>Conclusions: </strong>Our study identified a novel oncogenic epi-transcriptome axis involving NAT10-ac4C/eEF2-HMGB2, which plays a pivotal role in regulating HCC growth and metastasis. The drug Panobinostat validates the therapeutic potential of targeting this axis for HCC treatment.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1000,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1002/cac2.12595","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: N4-acetylcytidine (ac4C) represents a novel messenger RNA (mRNA) modification, and its associated acetyltransferase N-acetyltransferase 10 (NAT10) plays a crucial role in the initiation and progression of tumors by regulating mRNA functionality. However, its role in hepatocellular carcinoma (HCC) development and prognosis is largely unknown. This study aimed to elucidate the role of NAT10-mediated ac4C in HCC progression and provide a promising therapeutic approach.

Methods: The ac4C levels were evaluated by dot blot and ultra-performance liquid chromatography-tandem mass spectrometry with harvested HCC tissues. The expression of NAT10 was investigated using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemical staining across 91 cohorts of HCC patients. To explore the underlying mechanisms of NAT10-ac4C in HCC, we employed a comprehensive approach integrating acetylated RNA immunoprecipitation and sequencing, RNA sequencing and ribosome profiling analyses, along with RNA immunoprecipitation, RNA pull-down, mass spectrometry, and site-specific mutation analyses. The drug affinity responsive targets stability, cellular thermal shift assay, and surface plasmon resonance assays were performed to assess the specific binding of NAT10 and Panobinostat. Furthermore, the efficacy of targeting NAT10-ac4C for HCC treatment was elucidated through in vitro experiments using HCC cells and in vivo HCC mouse models.

Results: Our investigation revealed a significant increase in both the ac4C RNA level and NAT10 expression in HCC. Notably, elevated NAT10 expression was associated with poor outcomes in HCC patients. Functionally, silencing NAT10 suppressed HCC proliferation and metastasis in vitro and in vivo. Mechanistically, NAT10 stimulates the ac4C modification within the coding sequence (CDS) of high mobility group protein B2 (HMGB2), which subsequently enhances HMGB2 translation by facilitating eukaryotic elongation factor 2 (eEF2) binding to the ac4C sites on HMGB2 mRNA's CDS. Additionally, high-throughput compound library screening revealed Panobinostat as a potent inhibitor of NAT10-mediated ac4C modification. This inhibition significantly attenuated HCC growth and metastasis in both in vitro experiments using HCC cells and in vivo HCC mouse models.

Conclusions: Our study identified a novel oncogenic epi-transcriptome axis involving NAT10-ac4C/eEF2-HMGB2, which plays a pivotal role in regulating HCC growth and metastasis. The drug Panobinostat validates the therapeutic potential of targeting this axis for HCC treatment.

通过抑制 eEF2 介导的 HMGB2 mRNA 翻译,靶向 N4-乙酰胞嘧啶可抑制肝细胞癌的进展。
背景:N4-乙酰胞苷(ac4C)是一种新型的信使 RNA(mRNA)修饰,与之相关的乙酰转移酶 N-acetyltransferase 10(NAT10)通过调节 mRNA 的功能在肿瘤的发生和发展过程中起着至关重要的作用。然而,它在肝细胞癌(HCC)的发展和预后中的作用在很大程度上是未知的。本研究旨在阐明 NAT10 介导的 ac4C 在 HCC 进展中的作用,并提供一种有前景的治疗方法:方法:通过点印迹和超高效液相色谱-串联质谱法对采集的 HCC 组织中的 ac4C 水平进行评估。采用定量实时聚合酶链式反应、Western 印迹法和免疫组化染色法研究了 91 组 HCC 患者中 NAT10 的表达情况。为了探索 NAT10-ac4C 在 HCC 中的潜在机制,我们采用了一种综合方法,将乙酰化 RNA 免疫沉淀和测序、RNA 测序和核糖体图谱分析以及 RNA 免疫沉淀、RNA 拉取、质谱分析和位点特异性突变分析结合在一起。为了评估 NAT10 和 Panobinostat 的特异性结合,还进行了药物亲和力反应靶点稳定性、细胞热转移试验和表面等离子体共振试验。此外,通过使用 HCC 细胞的体外实验和体内 HCC 小鼠模型,阐明了靶向 NAT10-ac4C 治疗 HCC 的疗效:结果:我们的研究发现,HCC中的ac4C RNA水平和NAT10表达均有明显增加。值得注意的是,NAT10 表达的升高与 HCC 患者的不良预后有关。从功能上讲,沉默 NAT10 可抑制 HCC 在体外和体内的增殖和转移。从机理上讲,NAT10能刺激高迁移率基团蛋白B2(HMGB2)编码序列(CDS)中的ac4C修饰,随后通过促进真核延伸因子2(eEF2)与HMGB2 mRNA的CDS上的ac4C位点结合来增强HMGB2的翻译。此外,高通量化合物库筛选发现,Panobinostat 是 NAT10 介导的 ac4C 修饰的强效抑制剂。在使用 HCC 细胞进行的体外实验和体内 HCC 小鼠模型中,这种抑制都能明显减轻 HCC 的生长和转移:我们的研究发现了一个涉及 NAT10-ac4C/eEF2-HMGB2 的新型致癌外转录组轴,它在调控 HCC 生长和转移方面发挥着关键作用。药物 Panobinostat 验证了针对该轴治疗 HCC 的治疗潜力。
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
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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