neil3介导的有丝分裂碱基切除修复端粒氧化损伤可防止肝细胞癌衰老

H. Gad, Z. Zhenjun, Carlos Benítez-Buelga, K. Sanjiv, Huang Xiangwei, He Kang, Feng Mingxuan, Zhao Zhicong, U. W. Berglund, Xia Qiang, T. Helleday
{"title":"neil3介导的有丝分裂碱基切除修复端粒氧化损伤可防止肝细胞癌衰老","authors":"H. Gad, Z. Zhenjun, Carlos Benítez-Buelga, K. Sanjiv, Huang Xiangwei, He Kang, Feng Mingxuan, Zhao Zhicong, U. W. Berglund, Xia Qiang, T. Helleday","doi":"10.3390/iecc2021-09227","DOIUrl":null,"url":null,"abstract":"While the repair of DNA double-strand breaks is known to be confined to different phases of the cell cycle and differentially activated at telomeres, less is known of compartmentalisation of base excision repair (BER). Here, we report that Endonuclease VIII like protein 3 (NEIL3) relocates to telomeres following oxidative DNA damage specifically during mitosis and recruits the APE1 to damaged telomeres. Using META-FISH, we demonstrate that NEIL3, but not NEIL1 or NEIL2, is required to initiate base excision repair at oxidised telomeres in mitotic cells, a process dependent on APE1 and Polβ. Repetitive exposure of oxidizing damage in NEIL3 depleted cells induced chromatin bridges and damaged telomeres. Interestingly, we identify that NEIL3 is elevated in Hepatocellular carcinoma (HCC), the most common type of a liver cancer, which correlates with poor survival. We demonstrate that HCC cell lines (6/6) depend on NEIL3 catalytic activity for survival and prevention of senescence, which is not the case for non-transformed cells where NEIL3 is dispensable. In conclusion, we demonstrate a novel function for NEIL3 in repair of oxidative DNA damage at telomeres in mitosis which is important to prevent senescence of HCC. Furthermore, these data suggest NEIL3 could be a target for therapeutic intervention of HCC.","PeriodicalId":20534,"journal":{"name":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2021-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"NEIL3-mediated mitotic base excision repair of oxidative lesions at telomeres prevents senescence in hepatocellular carcinoma\",\"authors\":\"H. Gad, Z. Zhenjun, Carlos Benítez-Buelga, K. Sanjiv, Huang Xiangwei, He Kang, Feng Mingxuan, Zhao Zhicong, U. W. Berglund, Xia Qiang, T. Helleday\",\"doi\":\"10.3390/iecc2021-09227\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"While the repair of DNA double-strand breaks is known to be confined to different phases of the cell cycle and differentially activated at telomeres, less is known of compartmentalisation of base excision repair (BER). Here, we report that Endonuclease VIII like protein 3 (NEIL3) relocates to telomeres following oxidative DNA damage specifically during mitosis and recruits the APE1 to damaged telomeres. Using META-FISH, we demonstrate that NEIL3, but not NEIL1 or NEIL2, is required to initiate base excision repair at oxidised telomeres in mitotic cells, a process dependent on APE1 and Polβ. Repetitive exposure of oxidizing damage in NEIL3 depleted cells induced chromatin bridges and damaged telomeres. Interestingly, we identify that NEIL3 is elevated in Hepatocellular carcinoma (HCC), the most common type of a liver cancer, which correlates with poor survival. We demonstrate that HCC cell lines (6/6) depend on NEIL3 catalytic activity for survival and prevention of senescence, which is not the case for non-transformed cells where NEIL3 is dispensable. In conclusion, we demonstrate a novel function for NEIL3 in repair of oxidative DNA damage at telomeres in mitosis which is important to prevent senescence of HCC. Furthermore, these data suggest NEIL3 could be a target for therapeutic intervention of HCC.\",\"PeriodicalId\":20534,\"journal\":{\"name\":\"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2021-01-31\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.3390/iecc2021-09227\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Proceedings of The 1st International Electronic Conference on Cancers: Exploiting Cancer Vulnerability by Targeting the DNA Damage Response","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3390/iecc2021-09227","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

虽然DNA双链断裂的修复已知局限于细胞周期的不同阶段,并在端粒上被差异激活,但对碱基切除修复(BER)的区隔化知之甚少。在这里,我们报道了内切酶VIII样蛋白3 (NEIL3)在有丝分裂期间特异性地在DNA氧化损伤后重新定位到端粒,并将APE1招募到受损的端粒。使用META-FISH,我们证明了NEIL3,而不是NEIL1或NEIL2,是启动有丝分裂细胞氧化端粒碱基切除修复所必需的,这一过程依赖于APE1和Polβ。重复暴露于NEIL3缺失细胞的氧化损伤诱导染色质桥和端粒损伤。有趣的是,我们发现NEIL3在肝细胞癌(HCC)中升高,HCC是最常见的肝癌类型,与生存率低相关。我们证明,HCC细胞系(6/6)依赖NEIL3的催化活性来存活和预防衰老,而非转化细胞则不是这样,因为NEIL3是必不可少的。总之,我们证明了NEIL3在有丝分裂中修复端粒氧化DNA损伤的新功能,这对预防HCC衰老很重要。此外,这些数据表明NEIL3可能是HCC治疗干预的靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
NEIL3-mediated mitotic base excision repair of oxidative lesions at telomeres prevents senescence in hepatocellular carcinoma
While the repair of DNA double-strand breaks is known to be confined to different phases of the cell cycle and differentially activated at telomeres, less is known of compartmentalisation of base excision repair (BER). Here, we report that Endonuclease VIII like protein 3 (NEIL3) relocates to telomeres following oxidative DNA damage specifically during mitosis and recruits the APE1 to damaged telomeres. Using META-FISH, we demonstrate that NEIL3, but not NEIL1 or NEIL2, is required to initiate base excision repair at oxidised telomeres in mitotic cells, a process dependent on APE1 and Polβ. Repetitive exposure of oxidizing damage in NEIL3 depleted cells induced chromatin bridges and damaged telomeres. Interestingly, we identify that NEIL3 is elevated in Hepatocellular carcinoma (HCC), the most common type of a liver cancer, which correlates with poor survival. We demonstrate that HCC cell lines (6/6) depend on NEIL3 catalytic activity for survival and prevention of senescence, which is not the case for non-transformed cells where NEIL3 is dispensable. In conclusion, we demonstrate a novel function for NEIL3 in repair of oxidative DNA damage at telomeres in mitosis which is important to prevent senescence of HCC. Furthermore, these data suggest NEIL3 could be a target for therapeutic intervention of HCC.
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
自引率
0.00%
发文量
0
×
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