ZMIZ1 enhances ERα-dependent expression of E2F2 in breast cancer

IF 3.6 4区 医学 Q2 ENDOCRINOLOGY & METABOLISM Journal of molecular endocrinology Pub Date : 2024-04-01 DOI:10.1530/jme-23-0133
Weiye Zhao, Susanna F Rose, Ryan Blake, Aňze Godicelj, Amy E Cullen, Jack Stenning, Lucy Beevors, Marcel Gehrung, Sanjeev Kumar, Kamal Kishore, Ashley Sawle, Matthew Eldridge, Federico M Giorgi, Katherine S Bridge, Florian Markowetz, Andrew N Holding
{"title":"ZMIZ1 enhances ERα-dependent expression of E2F2 in breast cancer","authors":"Weiye Zhao, Susanna F Rose, Ryan Blake, Aňze Godicelj, Amy E Cullen, Jack Stenning, Lucy Beevors, Marcel Gehrung, Sanjeev Kumar, Kamal Kishore, Ashley Sawle, Matthew Eldridge, Federico M Giorgi, Katherine S Bridge, Florian Markowetz, Andrew N Holding","doi":"10.1530/jme-23-0133","DOIUrl":null,"url":null,"abstract":"<p>The Estrogen Receptor-alpha (ER) drives 75% of breast cancers. On activation, the ER recruits and assembles a 1-2 MDa transcriptionally active complex. These complexes can modulate tumour growth, and understanding the roles of individual proteins within these complexes can help identify new therapeutic targets. Here, we present the discovery of ER and ZMIZ1 within the same multi-protein assembly by quantitative proteomics, and validated by proximity ligation assay. We characterise ZMIZ1 function by demonstrating a significant decrease in the proliferation of ER-positive cancer cell lines. To establish a role for the ER-ZMIZ1 interaction, we measured the transcriptional changes in the estrogen response post-ZMIZ1 knockdown using an RNA-seq time-course over 24 hours. GSEA analysis of the ZMIZ1-knockdown data identified a specific delay in the response of estradiol-induced cell cycle genes. Integration of ENCODE data with our RNA-seq results identified that ER and ZMIZ1 both bind the promoter of E2F2. We therefore propose that ER and ZMIZ1 interact to enable the efficient estrogenic response at subset of cell cycle genes via a novel ZMIZ1-ER-E2F2 signalling axis. Finally, we show that high ZMIZ1 expression is predictive of worse patient outcome, ER and ZMIZ1 are co-expressed in breast cancer patients in TCGA and METABRIC, and the proteins are co-localised within the nuclei of tumours cell in patient biopsies. In conclusion, we establish that ZMIZ1 is a regulator of the estrogenic cell cycle response and provide evidence of the biological importance of the ER-ZMIZ1 interaction in ER-positive patient tumours, supporting potential clinical relevance.</p>","PeriodicalId":16570,"journal":{"name":"Journal of molecular endocrinology","volume":null,"pages":null},"PeriodicalIF":3.6000,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of molecular endocrinology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1530/jme-23-0133","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

Abstract

The Estrogen Receptor-alpha (ER) drives 75% of breast cancers. On activation, the ER recruits and assembles a 1-2 MDa transcriptionally active complex. These complexes can modulate tumour growth, and understanding the roles of individual proteins within these complexes can help identify new therapeutic targets. Here, we present the discovery of ER and ZMIZ1 within the same multi-protein assembly by quantitative proteomics, and validated by proximity ligation assay. We characterise ZMIZ1 function by demonstrating a significant decrease in the proliferation of ER-positive cancer cell lines. To establish a role for the ER-ZMIZ1 interaction, we measured the transcriptional changes in the estrogen response post-ZMIZ1 knockdown using an RNA-seq time-course over 24 hours. GSEA analysis of the ZMIZ1-knockdown data identified a specific delay in the response of estradiol-induced cell cycle genes. Integration of ENCODE data with our RNA-seq results identified that ER and ZMIZ1 both bind the promoter of E2F2. We therefore propose that ER and ZMIZ1 interact to enable the efficient estrogenic response at subset of cell cycle genes via a novel ZMIZ1-ER-E2F2 signalling axis. Finally, we show that high ZMIZ1 expression is predictive of worse patient outcome, ER and ZMIZ1 are co-expressed in breast cancer patients in TCGA and METABRIC, and the proteins are co-localised within the nuclei of tumours cell in patient biopsies. In conclusion, we establish that ZMIZ1 is a regulator of the estrogenic cell cycle response and provide evidence of the biological importance of the ER-ZMIZ1 interaction in ER-positive patient tumours, supporting potential clinical relevance.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
ZMIZ1 可增强乳腺癌中 ERα 依赖性 E2F2 的表达
75% 的乳腺癌是由雌激素受体-α(ER)引起的。雌激素受体激活后,会招募并组装一个 1-2 MDa 的转录活性复合物。这些复合物可以调节肿瘤的生长,了解这些复合物中各个蛋白质的作用有助于确定新的治疗靶点。在这里,我们通过定量蛋白质组学发现了ER和ZMIZ1在同一个多蛋白集合体中的作用,并通过邻近连接试验进行了验证。我们通过证明ER阳性癌细胞株的增殖显著下降来描述ZMIZ1的功能。为了确定ER-ZMIZ1相互作用的作用,我们使用RNA-seq时间序列测定了24小时内ZMIZ1敲除后雌激素反应的转录变化。对ZMIZ1敲除数据的GSEA分析发现,雌二醇诱导的细胞周期基因的反应出现了特定的延迟。将ENCODE数据与我们的RNA-seq结果整合后发现,ER和ZMIZ1都与E2F2的启动子结合。因此,我们认为ER和ZMIZ1相互作用,通过新的ZMIZ1-ER-E2F2信号轴在细胞周期基因子集上实现了有效的雌激素反应。最后,我们发现 ZMIZ1 的高表达预示着患者预后的恶化,在 TCGA 和 METABRIC 中,ER 和 ZMIZ1 在乳腺癌患者中共同表达,而且这两种蛋白在患者活检的肿瘤细胞核内共同定位。总之,我们证实 ZMIZ1 是雌激素细胞周期反应的调节因子,并提供了 ER-ZMIZ1 相互作用在 ER 阳性患者肿瘤中的生物学重要性的证据,支持其潜在的临床意义。
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 去求助
来源期刊
Journal of molecular endocrinology
Journal of molecular endocrinology 医学-内分泌学与代谢
CiteScore
6.90
自引率
0.00%
发文量
96
审稿时长
1 months
期刊介绍: The Journal of Molecular Endocrinology is an official journal of the Society for Endocrinology and is endorsed by the European Society of Endocrinology and the Endocrine Society of Australia. Journal of Molecular Endocrinology is a leading global journal that publishes original research articles and reviews. The journal focuses on molecular and cellular mechanisms in endocrinology, including: gene regulation, cell biology, signalling, mutations, transgenics, hormone-dependant cancers, nuclear receptors, and omics. Basic and pathophysiological studies at the molecule and cell level are considered, as well as human sample studies where this is the experimental model of choice. Technique studies including CRISPR or gene editing are also encouraged.
期刊最新文献
Increased capacity to maintain glucose homeostasis in a transgenic mouse expressing human but not mouse growth hormone with developing high-fat diet-related insulin resistance, hepatic steatosis and adipose dysfunction. Heterogeneous metabolic changes of brown and white adipose tissues are associated with metabolic adaptations in periparturient mice. LIF-STAT signaling in decidual cells: a possible role in embryo implantation and early pregnancy. ZMIZ1 enhances ERα-dependent expression of E2F2 in breast cancer circ_0134120: a new frontier in understanding postmenopausal osteoporosis pathogenesis
×
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
现在去查看 取消
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1