当前位置: X-MOL 学术Theranostics › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
YTHDF1-enhanced iron metabolism depends on TFRC m6A methylation
Theranostics ( IF 12.4 ) Pub Date : 2020-10-26 , DOI: 10.7150/thno.51231
Jing Ye 1 , Zhanggui Wang 2 , Xiaozhen Chen 3 , Xiaohua Jiang 1 , Zhihuai Dong 1 , Sunhong Hu 1 , Wenya Li 1 , Yuehui Liu 4 , Bing Liao 4 , Weidong Han 5 , Jiaying Shen 5 , Mang Xiao 1
Affiliation  

Background: Among head and neck squamous cell carcinomas (HNSCCs), hypopharyngeal squamous cell carcinoma (HPSCC) has the worst prognosis. Iron metabolism, which plays a crucial role in tumor progression, is mainly regulated by alterations to genes and post-transcriptional processes. The recent discovery of the N6-methyladenosine (m6A) modification has expanded the realm of previously undiscovered post-transcriptional gene regulation mechanisms in eukaryotes. Many studies have demonstrated that m6A methylation represents a distinct layer of epigenetic deregulation in carcinogenesis and tumor proliferation. However, the status of m6A modification and iron metabolism in HPSCC remains unknown./nMethods: Bioinformatics analysis, sample analysis, and transcriptome sequencing were performed to evaluate the correlation between m6A modification and iron metabolism. Iron metabolic and cell biological analyses were conducted to evaluate the effect of the m6A reader YTHDF1 on HPSCC proliferation and iron metabolism. Transcriptome-wide m6A-seq and RIP-seq data were mapped to explore the molecular mechanism of YTHDF1 function in HPSCC./nResults: YTHDF1 was found to be closely associated with ferritin levels and intratumoral iron concentrations in HPSCC patients at Sir Run Run Shaw Hospital. YTHDF1 induced-HPSCC tumorigenesis depends on iron metabolism in vivo in vitro. Mechanistically, YTHDF1 methyltransferase domain interacts with the 3'UTR and 5'UTR of TRFC mRNA, then further positively regulates translation of m6A-modified TFRC mRNA. Gain-of-function and loss-of-function analyses validated the finding showing that TFRC is a crucial target gene for YTHDF1-mediated increases in iron metabolism./nConclusion: YTHDF1 enhanced TFRC expression in HPSCC through an m6A-dependent mechanism. From a therapeutic perspective, targeting YTHDF1 and TFRC-mediated iron metabolism may be a promising strategy for HPSCC.

中文翻译:

YTHDF1 增强的铁代谢取决于 TFRC m6A 甲基化

背景:在头颈部鳞状细胞癌 (HNSCC) 中,下咽鳞状细胞癌 (HPSCC) 的预后最差。铁代谢在肿瘤进展中起关键作用,主要受基因改变和转录后过程的调节。最近发现的 N6-甲基腺苷 (m 6 A) 修饰扩展了真核生物中先前未发现的转录后基因调控机制的领域。许多研究表明,m 6 A 甲基化代表了致癌作用和肿瘤增殖中表观遗传失调的独特层。然而,HPSCC中 m 6 A 修饰和铁代谢的状态仍然未知。/n方法:进行生物信息学分析、样品分析和转录组测序以评估 m 6 A 修饰与铁代谢之间的相关性。进行铁代谢和细胞生物学分析以评估 m 6 A 阅读器 YTHDF1 对 HPSCC 增殖和铁代谢的影响。转录组范围内的 m 6 A-seq 和 RIP-seq 数据被映射以探索 YTHDF1 在 HPSCC 中功能的分子机制。/n结果:在 Sir Run 发现 YTHDF1 与 HPSCC 患者的铁蛋白水平和瘤内铁浓度密切相关逸夫医院。YTHDF1 诱导的 HPSCC 肿瘤发生依赖于体内 体外铁代谢. 从机制上讲,YTHDF1 甲基转移酶结构域与 TRFC mRNA 的 3'UTR 和 5'UTR 相互作用,然后进一步正向调节 m 6 A 修饰的 TFRC mRNA 的翻译。功能获得和功能丧失分析证实了这一发现,表明 TFRC 是 YTHDF1 介导的铁代谢增加的关键靶基因。/n结论: YTHDF1 通过 m 6 A 依赖性机制增强 HPSCC 中的 TFRC 表达. 从治疗的角度来看,靶向 YTHDF1 和 TFRC 介导的铁代谢可能是 HPSCC 的一种有前景的策略。
更新日期:2020-11-02
down
wechat
bug