AKT1调节UHRF1蛋白稳定性,促进前列腺癌对阿比特龙的抵抗。

IF 5.9 2区 医学 Q1 ONCOLOGY
Yongming Fu, Tuoyu Cao, Xiaorui Zou, Yubing Ye, Youhong Liu, Yuchong Peng, Tanggang Deng, Linglong Yin, Xiong Li
{"title":"AKT1调节UHRF1蛋白稳定性,促进前列腺癌对阿比特龙的抵抗。","authors":"Yongming Fu,&nbsp;Tuoyu Cao,&nbsp;Xiaorui Zou,&nbsp;Yubing Ye,&nbsp;Youhong Liu,&nbsp;Yuchong Peng,&nbsp;Tanggang Deng,&nbsp;Linglong Yin,&nbsp;Xiong Li","doi":"10.1038/s41389-022-00446-y","DOIUrl":null,"url":null,"abstract":"<p><p>Oncogenic activation of PI3K/AKT signaling pathway, together with epigenetic aberrations are the characters of castration-resistant prostate cancer (CRPC). UHRF1 as a key epigenetic regulator, plays a critical role in prostate cancer (PCa) development, and its expression is positively correlated with the degree of malignancy. In this present study we investigated the potential regulatory mechanism of AKT1 on UHRF1, and further validated the in vitro and in vivo anticancer efficacy of AKT phosphorylation inhibitor MK2206 in combination with abiraterone. Both UHRF1 and p-AKT aberrantly overexpressed in the abiraterone-resistant PCa cells. Further studies revealed that AKT1 protein interacts with UHRF1, and AKT1 directly phosphorylates UHRF1 via the site Thr-210. MK2206 induced UHRF1 protein degradation by inhibiting AKT1-induced UHRF1 phosphorylation, and then reduced the interaction between UHRF1 and deubiquitinase USP7, while promoted the interaction between UHRF1 and E3 ubiquitin protein ligase BTRC. MK2206 significantly promoted the sensitivity of abiraterone-refractory PCa cells and xenografts to abiraterone by decreasing UHRF1 protein level, and reversed the phenotype of NEPC, evently induced cellular senescence and cell apoptosis. Altogether, our present study for the first time revealed a novel molecular mechanism of abiraterone resistance through PI3K/AKT-UHRF1 pathway, and provided a novel therapeutic modality by targeting PI3K/AKT1 to promote the drug sensitivity of abiraterone in PCa patients.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9000,"publicationDate":"2023-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9807647/pdf/","citationCount":"3","resultStr":"{\"title\":\"AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.\",\"authors\":\"Yongming Fu,&nbsp;Tuoyu Cao,&nbsp;Xiaorui Zou,&nbsp;Yubing Ye,&nbsp;Youhong Liu,&nbsp;Yuchong Peng,&nbsp;Tanggang Deng,&nbsp;Linglong Yin,&nbsp;Xiong Li\",\"doi\":\"10.1038/s41389-022-00446-y\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Oncogenic activation of PI3K/AKT signaling pathway, together with epigenetic aberrations are the characters of castration-resistant prostate cancer (CRPC). UHRF1 as a key epigenetic regulator, plays a critical role in prostate cancer (PCa) development, and its expression is positively correlated with the degree of malignancy. In this present study we investigated the potential regulatory mechanism of AKT1 on UHRF1, and further validated the in vitro and in vivo anticancer efficacy of AKT phosphorylation inhibitor MK2206 in combination with abiraterone. Both UHRF1 and p-AKT aberrantly overexpressed in the abiraterone-resistant PCa cells. Further studies revealed that AKT1 protein interacts with UHRF1, and AKT1 directly phosphorylates UHRF1 via the site Thr-210. MK2206 induced UHRF1 protein degradation by inhibiting AKT1-induced UHRF1 phosphorylation, and then reduced the interaction between UHRF1 and deubiquitinase USP7, while promoted the interaction between UHRF1 and E3 ubiquitin protein ligase BTRC. MK2206 significantly promoted the sensitivity of abiraterone-refractory PCa cells and xenografts to abiraterone by decreasing UHRF1 protein level, and reversed the phenotype of NEPC, evently induced cellular senescence and cell apoptosis. Altogether, our present study for the first time revealed a novel molecular mechanism of abiraterone resistance through PI3K/AKT-UHRF1 pathway, and provided a novel therapeutic modality by targeting PI3K/AKT1 to promote the drug sensitivity of abiraterone in PCa patients.</p>\",\"PeriodicalId\":19489,\"journal\":{\"name\":\"Oncogenesis\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":5.9000,\"publicationDate\":\"2023-01-02\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9807647/pdf/\",\"citationCount\":\"3\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Oncogenesis\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1038/s41389-022-00446-y\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncogenesis","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41389-022-00446-y","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 3

摘要

PI3K/AKT信号通路的致瘤激活以及表观遗传畸变是去势抵抗性前列腺癌(CRPC)的特征。UHRF1作为一种关键的表观遗传调控因子,在前列腺癌(prostate cancer, PCa)的发生发展中起着至关重要的作用,其表达与恶性程度呈正相关。本研究探讨了AKT1对UHRF1的潜在调控机制,进一步验证了AKT磷酸化抑制剂MK2206联合阿比特龙在体外和体内的抗癌效果。UHRF1和p-AKT在阿比特龙耐药PCa细胞中均异常过表达。进一步的研究发现AKT1蛋白与UHRF1相互作用,AKT1通过位点Thr-210直接磷酸化UHRF1。MK2206通过抑制akt1诱导的UHRF1磷酸化诱导UHRF1蛋白降解,进而降低UHRF1与去泛素酶USP7的相互作用,促进UHRF1与E3泛素蛋白连接酶BTRC的相互作用。MK2206通过降低UHRF1蛋白水平,显著提高阿比特龙难愈性PCa细胞和异种移植物对阿比特龙的敏感性,逆转NEPC表型,诱导细胞衰老和细胞凋亡。综上所述,本研究首次通过PI3K/AKT-UHRF1途径揭示了阿比特龙耐药的新分子机制,并提供了一种以PI3K/AKT1为靶点促进阿比特龙在PCa患者药物敏感性的新治疗模式。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.

AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.

AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.

AKT1 regulates UHRF1 protein stability and promotes the resistance to abiraterone in prostate cancer.

Oncogenic activation of PI3K/AKT signaling pathway, together with epigenetic aberrations are the characters of castration-resistant prostate cancer (CRPC). UHRF1 as a key epigenetic regulator, plays a critical role in prostate cancer (PCa) development, and its expression is positively correlated with the degree of malignancy. In this present study we investigated the potential regulatory mechanism of AKT1 on UHRF1, and further validated the in vitro and in vivo anticancer efficacy of AKT phosphorylation inhibitor MK2206 in combination with abiraterone. Both UHRF1 and p-AKT aberrantly overexpressed in the abiraterone-resistant PCa cells. Further studies revealed that AKT1 protein interacts with UHRF1, and AKT1 directly phosphorylates UHRF1 via the site Thr-210. MK2206 induced UHRF1 protein degradation by inhibiting AKT1-induced UHRF1 phosphorylation, and then reduced the interaction between UHRF1 and deubiquitinase USP7, while promoted the interaction between UHRF1 and E3 ubiquitin protein ligase BTRC. MK2206 significantly promoted the sensitivity of abiraterone-refractory PCa cells and xenografts to abiraterone by decreasing UHRF1 protein level, and reversed the phenotype of NEPC, evently induced cellular senescence and cell apoptosis. Altogether, our present study for the first time revealed a novel molecular mechanism of abiraterone resistance through PI3K/AKT-UHRF1 pathway, and provided a novel therapeutic modality by targeting PI3K/AKT1 to promote the drug sensitivity of abiraterone in PCa patients.

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
Oncogenesis
Oncogenesis ONCOLOGY-
CiteScore
11.90
自引率
0.00%
发文量
70
审稿时长
26 weeks
期刊介绍: Oncogenesis is a peer-reviewed open access online journal that publishes full-length papers, reviews, and short communications exploring the molecular basis of cancer and related phenomena. It seeks to promote diverse and integrated areas of molecular biology, cell biology, oncology, and genetics.
×
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