KDM4/JMJD2组蛋白去甲基化酶抑制剂通过抑制AR和bmyb调节基因的表达来阻断前列腺肿瘤的生长。

Chemistry & biology Pub Date : 2015-09-17 Epub Date: 2015-09-10 DOI:10.1016/j.chembiol.2015.08.007
Lingling Duan, Ganesha Rai, Carlos Roggero, Qing-Jun Zhang, Qun Wei, Shi Hong Ma, Yunyun Zhou, John Santoyo, Elisabeth D Martinez, Guanghua Xiao, Ganesh V Raj, Ajit Jadhav, Anton Simeonov, David J Maloney, Josep Rizo, Jer-Tsong Hsieh, Zhi-Ping Liu
{"title":"KDM4/JMJD2组蛋白去甲基化酶抑制剂通过抑制AR和bmyb调节基因的表达来阻断前列腺肿瘤的生长。","authors":"Lingling Duan, Ganesha Rai, Carlos Roggero, Qing-Jun Zhang, Qun Wei, Shi Hong Ma, Yunyun Zhou, John Santoyo, Elisabeth D Martinez, Guanghua Xiao, Ganesh V Raj, Ajit Jadhav, Anton Simeonov, David J Maloney, Josep Rizo, Jer-Tsong Hsieh, Zhi-Ping Liu","doi":"10.1016/j.chembiol.2015.08.007","DOIUrl":null,"url":null,"abstract":"<p><p>Histone lysine demethylase KDM4/JMJD2s are overexpressed in many human tumors including prostate cancer (PCa). KDM4s are co-activators of androgen receptor (AR) and are thus potential therapeutic targets. Yet to date few KDM4 inhibitors that have anti-prostate tumor activity in vivo have been developed. Here, we report the anti-tumor growth effect and molecular mechanisms of three novel KDM4 inhibitors (A1, I9, and B3). These inhibitors repressed the transcription of both AR and BMYB-regulated genes. Compound B3 is highly selective for a variety of cancer cell lines including PC3 cells that lack AR. B3 inhibited the in vivo growth of tumors derived from PC3 cells and ex vivo human PCa explants. We identified a novel mechanism by which KDM4B activates the transcription of Polo-like kinase 1 (PLK1). B3 blocked the binding of KDM4B to the PLK1 promoter. Our studies suggest a potential mechanism-based therapeutic strategy for PCa and tumors with elevated KDM4B/PLK1 expression.</p>","PeriodicalId":9772,"journal":{"name":"Chemistry & biology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2015-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4578295/pdf/nihms719015.pdf","citationCount":"0","resultStr":"{\"title\":\"KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes.\",\"authors\":\"Lingling Duan, Ganesha Rai, Carlos Roggero, Qing-Jun Zhang, Qun Wei, Shi Hong Ma, Yunyun Zhou, John Santoyo, Elisabeth D Martinez, Guanghua Xiao, Ganesh V Raj, Ajit Jadhav, Anton Simeonov, David J Maloney, Josep Rizo, Jer-Tsong Hsieh, Zhi-Ping Liu\",\"doi\":\"10.1016/j.chembiol.2015.08.007\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Histone lysine demethylase KDM4/JMJD2s are overexpressed in many human tumors including prostate cancer (PCa). KDM4s are co-activators of androgen receptor (AR) and are thus potential therapeutic targets. Yet to date few KDM4 inhibitors that have anti-prostate tumor activity in vivo have been developed. Here, we report the anti-tumor growth effect and molecular mechanisms of three novel KDM4 inhibitors (A1, I9, and B3). These inhibitors repressed the transcription of both AR and BMYB-regulated genes. Compound B3 is highly selective for a variety of cancer cell lines including PC3 cells that lack AR. B3 inhibited the in vivo growth of tumors derived from PC3 cells and ex vivo human PCa explants. We identified a novel mechanism by which KDM4B activates the transcription of Polo-like kinase 1 (PLK1). B3 blocked the binding of KDM4B to the PLK1 promoter. Our studies suggest a potential mechanism-based therapeutic strategy for PCa and tumors with elevated KDM4B/PLK1 expression.</p>\",\"PeriodicalId\":9772,\"journal\":{\"name\":\"Chemistry & biology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2015-09-17\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4578295/pdf/nihms719015.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Chemistry & biology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1016/j.chembiol.2015.08.007\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2015/9/10 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Chemistry & biology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1016/j.chembiol.2015.08.007","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2015/9/10 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

组蛋白赖氨酸去甲基化酶KDM4/JMJD2s在包括前列腺癌(PCa)在内的许多人类肿瘤中过表达。kdm4是雄激素受体(AR)的共激活剂,因此是潜在的治疗靶点。然而,迄今为止,很少有KDM4抑制剂在体内具有抗前列腺肿瘤活性。在这里,我们报道了三种新型KDM4抑制剂(A1, I9和B3)的抗肿瘤生长作用和分子机制。这些抑制剂抑制AR和bmyb调节基因的转录。化合物B3对包括缺乏AR的PC3细胞在内的多种癌细胞系具有高度选择性。B3抑制来自PC3细胞和离体人PCa外植体的肿瘤的体内生长。我们确定了KDM4B激活polo样激酶1 (PLK1)转录的新机制。B3阻断KDM4B与PLK1启动子的结合。我们的研究为前列腺癌和KDM4B/PLK1表达升高的肿瘤提供了一种潜在的基于机制的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes.

KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes.

KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes.

KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes.

Histone lysine demethylase KDM4/JMJD2s are overexpressed in many human tumors including prostate cancer (PCa). KDM4s are co-activators of androgen receptor (AR) and are thus potential therapeutic targets. Yet to date few KDM4 inhibitors that have anti-prostate tumor activity in vivo have been developed. Here, we report the anti-tumor growth effect and molecular mechanisms of three novel KDM4 inhibitors (A1, I9, and B3). These inhibitors repressed the transcription of both AR and BMYB-regulated genes. Compound B3 is highly selective for a variety of cancer cell lines including PC3 cells that lack AR. B3 inhibited the in vivo growth of tumors derived from PC3 cells and ex vivo human PCa explants. We identified a novel mechanism by which KDM4B activates the transcription of Polo-like kinase 1 (PLK1). B3 blocked the binding of KDM4B to the PLK1 promoter. Our studies suggest a potential mechanism-based therapeutic strategy for PCa and tumors with elevated KDM4B/PLK1 expression.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Chemistry & biology
Chemistry & biology 生物-生化与分子生物学
自引率
0.00%
发文量
0
审稿时长
4-8 weeks
×
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