Epigenetic reader ZMYND11 noncanonical function restricts HNRNPA1-mediated stress granule formation and oncogenic activity

IF 40.8 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY
Cheng Lian, Chunyi Zhang, Pan Tian, Qilong Tan, Yu Wei, Zixian Wang, Qin Zhang, Qixiang Zhang, Mengjie Zhong, Li-Quan Zhou, Xisong Ke, Huabing Zhang, Yao Zhu, Zhenfei Li, Jingdong Cheng, Gong-Hong Wei
{"title":"Epigenetic reader ZMYND11 noncanonical function restricts HNRNPA1-mediated stress granule formation and oncogenic activity","authors":"Cheng Lian, Chunyi Zhang, Pan Tian, Qilong Tan, Yu Wei, Zixian Wang, Qin Zhang, Qixiang Zhang, Mengjie Zhong, Li-Quan Zhou, Xisong Ke, Huabing Zhang, Yao Zhu, Zhenfei Li, Jingdong Cheng, Gong-Hong Wei","doi":"10.1038/s41392-024-01961-7","DOIUrl":null,"url":null,"abstract":"<p>Epigenetic readers frequently affect gene regulation, correlate with disease prognosis, and hold significant potential as therapeutic targets for cancer. Zinc finger MYND-type containing 11 (ZMYND11) is notably recognized for reading the epigenetic marker H3.3K36me3; however, its broader functions and mechanisms of action in cancer remain underexplored. Here, we report that ZMYND11 downregulation is prevalent across various cancers and profoundly correlates with poorer outcomes in prostate cancer patients. Depletion of ZMYND11 promotes tumor cell growth, migration, and invasion in vitro, as well as tumor formation and metastasis in vivo. Mechanistically, we discover that ZMYND11 exhibits tumor suppressive roles by recognizing arginine-194-methylated HNRNPA1 dependent on its MYND domain, thereby retaining HNRNPA1 in the nucleus and preventing the formation of stress granules in the cytoplasm. Furthermore, ZMYND11 counteracts the HNRNPA1-driven increase in the PKM2/PKM1 ratio, thus mitigating the aggressive tumor phenotype promoted by PKM2. Remarkably, ZMYND11 recognition of HNRNPA1 can be disrupted by pharmaceutical inhibition of the arginine methyltransferase PRMT5. Tumors with low ZMYND11 expression show sensitivity to PRMT5 inhibitors. Taken together, our findings uncover a previously unexplored noncanonical role of ZMYND11 as a nonhistone methylation reader and underscore the critical importance of arginine methylation in the ZMYND11-HNRNPA1 interaction for restraining tumor progression, thereby proposing novel therapeutic targets and potential biomarkers for cancer treatment.</p>","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":null,"pages":null},"PeriodicalIF":40.8000,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Signal Transduction and Targeted Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41392-024-01961-7","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Epigenetic readers frequently affect gene regulation, correlate with disease prognosis, and hold significant potential as therapeutic targets for cancer. Zinc finger MYND-type containing 11 (ZMYND11) is notably recognized for reading the epigenetic marker H3.3K36me3; however, its broader functions and mechanisms of action in cancer remain underexplored. Here, we report that ZMYND11 downregulation is prevalent across various cancers and profoundly correlates with poorer outcomes in prostate cancer patients. Depletion of ZMYND11 promotes tumor cell growth, migration, and invasion in vitro, as well as tumor formation and metastasis in vivo. Mechanistically, we discover that ZMYND11 exhibits tumor suppressive roles by recognizing arginine-194-methylated HNRNPA1 dependent on its MYND domain, thereby retaining HNRNPA1 in the nucleus and preventing the formation of stress granules in the cytoplasm. Furthermore, ZMYND11 counteracts the HNRNPA1-driven increase in the PKM2/PKM1 ratio, thus mitigating the aggressive tumor phenotype promoted by PKM2. Remarkably, ZMYND11 recognition of HNRNPA1 can be disrupted by pharmaceutical inhibition of the arginine methyltransferase PRMT5. Tumors with low ZMYND11 expression show sensitivity to PRMT5 inhibitors. Taken together, our findings uncover a previously unexplored noncanonical role of ZMYND11 as a nonhistone methylation reader and underscore the critical importance of arginine methylation in the ZMYND11-HNRNPA1 interaction for restraining tumor progression, thereby proposing novel therapeutic targets and potential biomarkers for cancer treatment.

Abstract Image

表观遗传读者 ZMYND11 的非规范功能限制了 HNRNPA1 介导的应激颗粒形成和致癌活性
表观遗传读取器经常影响基因调控,与疾病预后相关,并具有作为癌症治疗靶点的巨大潜力。锌指 MYND 型含 11(ZMYND11)被公认为可读取表观遗传标记 H3.3K36me3;然而,它在癌症中更广泛的功能和作用机制仍未得到充分探索。在此,我们报告了 ZMYND11 的下调在各种癌症中普遍存在,并且与前列腺癌患者的较差预后密切相关。ZMYND11 的耗竭会促进体外肿瘤细胞的生长、迁移和侵袭,以及体内肿瘤的形成和转移。从机理上讲,我们发现 ZMYND11 依赖于其 MYND 结构域识别精氨酸-194 甲基化的 HNRNPA1,从而将 HNRNPA1 保留在细胞核中,阻止细胞质中应激颗粒的形成,从而发挥抑制肿瘤的作用。此外,ZMYND11 还能抵消 HNRNPA1 驱动的 PKM2/PKM1 比率的增加,从而减轻 PKM2 促进的侵袭性肿瘤表型。值得注意的是,ZMYND11 对 HNRNPA1 的识别可被精氨酸甲基转移酶 PRMT5 的药物抑制所破坏。ZMYND11 低表达的肿瘤对 PRMT5 抑制剂敏感。总之,我们的研究结果揭示了 ZMYND11 作为非组蛋白甲基化阅读器的一种以前未曾探索过的非典型作用,并强调了精氨酸甲基化在 ZMYND11-HNRNPA1 相互作用中对抑制肿瘤进展的至关重要性,从而为癌症治疗提出了新的治疗靶点和潜在的生物标记物。
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
Signal Transduction and Targeted Therapy
Signal Transduction and Targeted Therapy Biochemistry, Genetics and Molecular Biology-Genetics
CiteScore
44.50
自引率
1.50%
发文量
384
审稿时长
5 weeks
期刊介绍: Signal Transduction and Targeted Therapy is an open access journal that focuses on timely publication of cutting-edge discoveries and advancements in basic science and clinical research related to signal transduction and targeted therapy. Scope: The journal covers research on major human diseases, including, but not limited to: Cancer,Cardiovascular diseases,Autoimmune diseases,Nervous system diseases.
×
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