The chromatin remodeler DEK promotes proliferation of mammary epithelium and is associated with H3K27me3 epigenetic modifications

Megan Johnstone, Ashley Leck, Taylor E Lange, Katherine E Wilcher, Miranda S Shephard, Aditi Paranjpe, Sophia Schutte, Susanne I Wells, Ferdinand Kappes, Nathan Salomonis, Lisa M Privette Vinnedge
{"title":"The chromatin remodeler DEK promotes proliferation of mammary epithelium and is associated with H3K27me3 epigenetic modifications","authors":"Megan Johnstone, Ashley Leck, Taylor E Lange, Katherine E Wilcher, Miranda S Shephard, Aditi Paranjpe, Sophia Schutte, Susanne I Wells, Ferdinand Kappes, Nathan Salomonis, Lisa M Privette Vinnedge","doi":"10.1101/2024.09.09.612116","DOIUrl":null,"url":null,"abstract":"The DEK chromatin remodeling protein was previously shown to confer oncogenic phenotypes to human and mouse mammary epithelial cells using in vitro and knockout mouse models. However, its functional role in normal mammary gland epithelium remained unexplored. We developed two novel mouse models to study the role of Dek in normal mammary gland biology in vivo. Mammary gland-specific Dek over-expression in mice resulted in hyperproliferation of cells that visually resembled alveolar cells, and a transcriptional profile that indicated increased expression of cell cycle, mammary stem/progenitor, and lactation-associated genes. Conversely, Dek knockout mice exhibited an alveologenesis or lactation defect, resulting in dramatically reduced pup survival. Analysis of previously published single-cell RNA-sequencing of mouse mammary glands revealed that Dek is most highly expressed in mammary stem cells and alveolar progenitor cells, and to a lesser extent in basal epithelial cells, supporting the observed phenotypes. Mechanistically, we discovered that Dek is a modifier of Ezh2 methyltransferase activity, upregulating the levels of histone H3 trimethylation on lysine 27 (H3K27me3) to control gene transcription. Combined, this work indicates that Dek promotes proliferation of mammary epithelial cells via cell cycle deregulation. Furthermore, we report a novel function for Dek in alveologenesis and histone H3 K27 trimethylation.","PeriodicalId":501269,"journal":{"name":"bioRxiv - Developmental Biology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"bioRxiv - Developmental Biology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.09.09.612116","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

The DEK chromatin remodeling protein was previously shown to confer oncogenic phenotypes to human and mouse mammary epithelial cells using in vitro and knockout mouse models. However, its functional role in normal mammary gland epithelium remained unexplored. We developed two novel mouse models to study the role of Dek in normal mammary gland biology in vivo. Mammary gland-specific Dek over-expression in mice resulted in hyperproliferation of cells that visually resembled alveolar cells, and a transcriptional profile that indicated increased expression of cell cycle, mammary stem/progenitor, and lactation-associated genes. Conversely, Dek knockout mice exhibited an alveologenesis or lactation defect, resulting in dramatically reduced pup survival. Analysis of previously published single-cell RNA-sequencing of mouse mammary glands revealed that Dek is most highly expressed in mammary stem cells and alveolar progenitor cells, and to a lesser extent in basal epithelial cells, supporting the observed phenotypes. Mechanistically, we discovered that Dek is a modifier of Ezh2 methyltransferase activity, upregulating the levels of histone H3 trimethylation on lysine 27 (H3K27me3) to control gene transcription. Combined, this work indicates that Dek promotes proliferation of mammary epithelial cells via cell cycle deregulation. Furthermore, we report a novel function for Dek in alveologenesis and histone H3 K27 trimethylation.
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
染色质重塑剂 DEK 能促进乳腺上皮细胞的增殖,并与 H3K27me3 表观遗传修饰有关
以前曾利用体外模型和基因敲除小鼠模型证明,DEK 染色质重塑蛋白可赋予人类和小鼠乳腺上皮细胞致癌表型。然而,它在正常乳腺上皮细胞中的功能作用仍有待探索。我们开发了两种新型小鼠模型来研究 Dek 在体内正常乳腺生物学中的作用。小鼠乳腺特异性 Dek 过度表达会导致细胞过度增殖,在视觉上与肺泡细胞相似,转录谱显示细胞周期、乳腺干/祖细胞和泌乳相关基因的表达增加。相反,Dek基因敲除小鼠表现出腺泡生成或泌乳缺陷,导致幼鼠存活率急剧下降。对先前发表的小鼠乳腺单细胞RNA序列分析表明,Dek在乳腺干细胞和腺泡祖细胞中的表达量最高,在基底上皮细胞中的表达量较低,这支持了观察到的表型。从机理上讲,我们发现Dek是Ezh2甲基转移酶活性的调节剂,能上调组蛋白H3赖氨酸27上的三甲基化(H3K27me3)水平,从而控制基因转录。综上所述,这项研究表明,Dek 通过细胞周期失调促进了乳腺上皮细胞的增殖。此外,我们还报告了 Dek 在腺泡生成和组蛋白 H3 K27 三甲基化中的新功能。
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
自引率
0.00%
发文量
0
期刊最新文献
The pericardium forms as a distinct structure during heart formation Centralspindlin promotes C. elegans anchor cell specification, vulva induction and morphogenesis Human macula formation involves two waves of retinoic acid signaling suppression via CYP26A1 regulating cell cycle exit and cone specification Single Cell Profiling in the Sox10Dom/+ Hirschsprung Mouse Implicates Hoxa6 in Enteric Neuron Lineage Allocation Mylpf dosage is proportionate to fast-twitch myofibril size in the zebrafish embryo
×
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
×
提示
确定
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