低剂量电离辐射通过非同源末端连接途径诱导RET/PTC1重排,从而引发甲状腺癌。

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL MedComm Pub Date : 2024-08-12 DOI:10.1002/mco2.690
Yuhao Liu, Jiaojiao Zhu, Shenghui Zhou, Yifan Hou, Ziyan Yan, Xingkun Ao, Ping Wang, Lin Zhou, Huixi Chen, Xinxin Liang, Hua Guan, Shanshan Gao, Dafei Xie, Yongqing Gu, Ping-Kun Zhou
{"title":"低剂量电离辐射通过非同源末端连接途径诱导RET/PTC1重排,从而引发甲状腺癌。","authors":"Yuhao Liu,&nbsp;Jiaojiao Zhu,&nbsp;Shenghui Zhou,&nbsp;Yifan Hou,&nbsp;Ziyan Yan,&nbsp;Xingkun Ao,&nbsp;Ping Wang,&nbsp;Lin Zhou,&nbsp;Huixi Chen,&nbsp;Xinxin Liang,&nbsp;Hua Guan,&nbsp;Shanshan Gao,&nbsp;Dafei Xie,&nbsp;Yongqing Gu,&nbsp;Ping-Kun Zhou","doi":"10.1002/mco2.690","DOIUrl":null,"url":null,"abstract":"<p>Thyroid cancer incidence increases worldwide annually, primarily due to factors such as ionizing radiation (IR), iodine intake, and genetics. Papillary carcinoma of the thyroid (PTC) accounts for about 80% of thyroid cancer cases. RET/PTC1 (coiled-coil domain containing 6 [CCDC6]-rearranged during transfection) rearrangement is a distinctive feature in over 70% of thyroid cancers who exposed to low doses of IR in Chernobyl and Hiroshima‒Nagasaki atomic bombings. This study aims to elucidate mechanism between RET/PTC1 rearrangement and IR in PTC. N-thy-ori-3-1 cells were subjected to varying doses of IR (2/1/0.5/0.2/0.1/0.05 Gy) of IR at different days, and result showed low-dose IR-induced RET/PTC1 rearrangement in a dose-dependent manner. RET/PTC1 has been observed to promote PTC both in vivo and in vitro. To delineate the role of different DNA repair pathways, SCR7, RI-1, and Olaparib were employed to inhibit non-homologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ), respectively. Notably, inhibiting NHEJ enhanced HR repair efficiency and reduced IR-induced RET/PTC1 rearrangement. Conversely, inhibiting HR increased NHEJ repair efficiency and subsequent RET/PTC1 rearrangement. The MMEJ did not show a markable role in this progress. Additionally, inhibiting DNA-dependent protein kinase catalytic subunit (DNA-PKcs) decreased the efficiency of NHEJ and thus reduced IR-induced RET/PTC1 rearrangement. To conclude, the data suggest that NHEJ, rather than HR or MMEJ, is the critical cause of IR-induced RET/PTC1 rearrangement. Targeting DNA-PKcs to inhibit the NHEJ has emerged as a promising therapeutic strategy for addressing IR-induced RET/PTC1 rearrangement in PTC.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":null,"pages":null},"PeriodicalIF":10.7000,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11318340/pdf/","citationCount":"0","resultStr":"{\"title\":\"Low-dose ionizing radiation-induced RET/PTC1 rearrangement via the non-homologous end joining pathway to drive thyroid cancer\",\"authors\":\"Yuhao Liu,&nbsp;Jiaojiao Zhu,&nbsp;Shenghui Zhou,&nbsp;Yifan Hou,&nbsp;Ziyan Yan,&nbsp;Xingkun Ao,&nbsp;Ping Wang,&nbsp;Lin Zhou,&nbsp;Huixi Chen,&nbsp;Xinxin Liang,&nbsp;Hua Guan,&nbsp;Shanshan Gao,&nbsp;Dafei Xie,&nbsp;Yongqing Gu,&nbsp;Ping-Kun Zhou\",\"doi\":\"10.1002/mco2.690\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Thyroid cancer incidence increases worldwide annually, primarily due to factors such as ionizing radiation (IR), iodine intake, and genetics. Papillary carcinoma of the thyroid (PTC) accounts for about 80% of thyroid cancer cases. RET/PTC1 (coiled-coil domain containing 6 [CCDC6]-rearranged during transfection) rearrangement is a distinctive feature in over 70% of thyroid cancers who exposed to low doses of IR in Chernobyl and Hiroshima‒Nagasaki atomic bombings. This study aims to elucidate mechanism between RET/PTC1 rearrangement and IR in PTC. N-thy-ori-3-1 cells were subjected to varying doses of IR (2/1/0.5/0.2/0.1/0.05 Gy) of IR at different days, and result showed low-dose IR-induced RET/PTC1 rearrangement in a dose-dependent manner. RET/PTC1 has been observed to promote PTC both in vivo and in vitro. To delineate the role of different DNA repair pathways, SCR7, RI-1, and Olaparib were employed to inhibit non-homologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ), respectively. Notably, inhibiting NHEJ enhanced HR repair efficiency and reduced IR-induced RET/PTC1 rearrangement. Conversely, inhibiting HR increased NHEJ repair efficiency and subsequent RET/PTC1 rearrangement. The MMEJ did not show a markable role in this progress. Additionally, inhibiting DNA-dependent protein kinase catalytic subunit (DNA-PKcs) decreased the efficiency of NHEJ and thus reduced IR-induced RET/PTC1 rearrangement. To conclude, the data suggest that NHEJ, rather than HR or MMEJ, is the critical cause of IR-induced RET/PTC1 rearrangement. Targeting DNA-PKcs to inhibit the NHEJ has emerged as a promising therapeutic strategy for addressing IR-induced RET/PTC1 rearrangement in PTC.</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2024-08-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11318340/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.690\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.690","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

甲状腺癌的发病率在全球范围内逐年上升,这主要是由于电离辐射(IR)、碘摄入量和遗传等因素造成的。甲状腺乳头状癌(PTC)约占甲状腺癌病例的 80%。在切尔诺贝利核电站和广岛长崎原子弹爆炸中暴露于低剂量红外辐射的甲状腺癌患者中,70%以上都存在RET/PTC1(含盘旋线圈结构域6 [CCDC6]-转染时重排)重排这一显著特征。本研究旨在阐明PTC中RET/PTC1重排与IR之间的机制。将 N-thy-ori-3-1 细胞置于不同剂量的红外(2/1/0.5/0.2/0.1/0.05 Gy)照射下不同天数,结果显示低剂量红外诱导的 RET/PTC1 重排呈剂量依赖性。据观察,RET/PTC1在体内和体外均可促进PTC的发生。为了明确不同DNA修复途径的作用,研究人员采用SCR7、RI-1和奥拉帕利分别抑制非同源末端连接(NHEJ)、同源重组(HR)和微同源介导的末端连接(MMEJ)。值得注意的是,抑制 NHEJ 可提高 HR 修复效率,减少红外诱导的 RET/PTC1 重排。相反,抑制 HR 则会提高 NHEJ 的修复效率和随后的 RET/PTC1 重排。在这一过程中,MMEJ 没有显示出明显的作用。此外,抑制 DNA 依赖性蛋白激酶催化亚基(DNA-PKcs)会降低 NHEJ 的效率,从而减少 IR 诱导的 RET/PTC1 重排。总之,这些数据表明,NHEJ 而不是 HR 或 MMEJ 是 IR 诱导 RET/PTC1 重排的关键原因。以 DNA-PKcs 为靶点抑制 NHEJ 已成为解决 IR 诱导的 PTC RET/PTC1 重排的一种很有前景的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Low-dose ionizing radiation-induced RET/PTC1 rearrangement via the non-homologous end joining pathway to drive thyroid cancer

Thyroid cancer incidence increases worldwide annually, primarily due to factors such as ionizing radiation (IR), iodine intake, and genetics. Papillary carcinoma of the thyroid (PTC) accounts for about 80% of thyroid cancer cases. RET/PTC1 (coiled-coil domain containing 6 [CCDC6]-rearranged during transfection) rearrangement is a distinctive feature in over 70% of thyroid cancers who exposed to low doses of IR in Chernobyl and Hiroshima‒Nagasaki atomic bombings. This study aims to elucidate mechanism between RET/PTC1 rearrangement and IR in PTC. N-thy-ori-3-1 cells were subjected to varying doses of IR (2/1/0.5/0.2/0.1/0.05 Gy) of IR at different days, and result showed low-dose IR-induced RET/PTC1 rearrangement in a dose-dependent manner. RET/PTC1 has been observed to promote PTC both in vivo and in vitro. To delineate the role of different DNA repair pathways, SCR7, RI-1, and Olaparib were employed to inhibit non-homologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ), respectively. Notably, inhibiting NHEJ enhanced HR repair efficiency and reduced IR-induced RET/PTC1 rearrangement. Conversely, inhibiting HR increased NHEJ repair efficiency and subsequent RET/PTC1 rearrangement. The MMEJ did not show a markable role in this progress. Additionally, inhibiting DNA-dependent protein kinase catalytic subunit (DNA-PKcs) decreased the efficiency of NHEJ and thus reduced IR-induced RET/PTC1 rearrangement. To conclude, the data suggest that NHEJ, rather than HR or MMEJ, is the critical cause of IR-induced RET/PTC1 rearrangement. Targeting DNA-PKcs to inhibit the NHEJ has emerged as a promising therapeutic strategy for addressing IR-induced RET/PTC1 rearrangement in PTC.

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
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
期刊最新文献
Copper homeostasis and cuproptosis in health and disease Liver cirrhosis: molecular mechanisms and therapeutic interventions High serum magnesium level is associated with increased mortality in patients with sepsis: an international, multicenter retrospective study N1-methylpseudouridine modification level correlates with protein expression, immunogenicity, and stability of mRNA Issue Information
×
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