CBLC通过泛素化和CTTN降解抑制乳腺癌细胞的增殖和转移。

IF 2.6 4区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY
Weiwei Li, Ting Lei, Xiaoyu Song, Chun Deng, Jingrun Lu, Wenwu Zhang, Zhenzhan Kuang, Yongyin He, Quan Zhou, Zhaoxun Luo, Fei Mo, Hanlin Yang, Jianfeng Hang, Bin Xiao, Linhai Li
{"title":"CBLC通过泛素化和CTTN降解抑制乳腺癌细胞的增殖和转移。","authors":"Weiwei Li,&nbsp;Ting Lei,&nbsp;Xiaoyu Song,&nbsp;Chun Deng,&nbsp;Jingrun Lu,&nbsp;Wenwu Zhang,&nbsp;Zhenzhan Kuang,&nbsp;Yongyin He,&nbsp;Quan Zhou,&nbsp;Zhaoxun Luo,&nbsp;Fei Mo,&nbsp;Hanlin Yang,&nbsp;Jianfeng Hang,&nbsp;Bin Xiao,&nbsp;Linhai Li","doi":"10.1080/10799893.2022.2116049","DOIUrl":null,"url":null,"abstract":"<p><p>The E3 ubiquitin ligase is an important regulator of cell signaling and proteostasis and is tightly controlled in many diseases, including cancer. Our study aimed to investigate the biological role of the E3 ubiquitin ligase CBLC in breast cancer and elucidate the specific mechanistic network underlying CBLC-mediated target substrate degradation, cell proliferation and metastasis. Here, we showed that CBLC expression was higher in breast cancer tissues and cells than that in normal tissues and cells. Higher expression of CBLC predicted a better prognosis for breast cancer patients. CBLC inhibited the proliferation, migration and invasion of breast cancer cells. Co-IP and immunofluorescence co-localization assays demonstrated that CBLC interacted with CTTN in the cytoplasm. CBLC promoted the degradation of CTTN through the ubiquitin-proteasome pathway without affecting its mRNA level. The inhibitory effect of CBLC on breast cancer cell proliferation, migration and invasion could partly be reversed by CTTN. Taken together, our study clarified the biological role of CBLC as a tumor suppressor and discovered its functional substrate, providing a molecular basis for CBLC/CTTN as a potential therapeutic target in breast cancer.</p>","PeriodicalId":16962,"journal":{"name":"Journal of Receptors and Signal Transduction","volume":null,"pages":null},"PeriodicalIF":2.6000,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"1","resultStr":"{\"title\":\"CBLC inhibits the proliferation and metastasis of breast cancer cells via ubiquitination and degradation of CTTN.\",\"authors\":\"Weiwei Li,&nbsp;Ting Lei,&nbsp;Xiaoyu Song,&nbsp;Chun Deng,&nbsp;Jingrun Lu,&nbsp;Wenwu Zhang,&nbsp;Zhenzhan Kuang,&nbsp;Yongyin He,&nbsp;Quan Zhou,&nbsp;Zhaoxun Luo,&nbsp;Fei Mo,&nbsp;Hanlin Yang,&nbsp;Jianfeng Hang,&nbsp;Bin Xiao,&nbsp;Linhai Li\",\"doi\":\"10.1080/10799893.2022.2116049\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>The E3 ubiquitin ligase is an important regulator of cell signaling and proteostasis and is tightly controlled in many diseases, including cancer. Our study aimed to investigate the biological role of the E3 ubiquitin ligase CBLC in breast cancer and elucidate the specific mechanistic network underlying CBLC-mediated target substrate degradation, cell proliferation and metastasis. Here, we showed that CBLC expression was higher in breast cancer tissues and cells than that in normal tissues and cells. Higher expression of CBLC predicted a better prognosis for breast cancer patients. CBLC inhibited the proliferation, migration and invasion of breast cancer cells. Co-IP and immunofluorescence co-localization assays demonstrated that CBLC interacted with CTTN in the cytoplasm. CBLC promoted the degradation of CTTN through the ubiquitin-proteasome pathway without affecting its mRNA level. The inhibitory effect of CBLC on breast cancer cell proliferation, migration and invasion could partly be reversed by CTTN. Taken together, our study clarified the biological role of CBLC as a tumor suppressor and discovered its functional substrate, providing a molecular basis for CBLC/CTTN as a potential therapeutic target in breast cancer.</p>\",\"PeriodicalId\":16962,\"journal\":{\"name\":\"Journal of Receptors and Signal Transduction\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.6000,\"publicationDate\":\"2022-12-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"1\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of Receptors and Signal Transduction\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1080/10799893.2022.2116049\",\"RegionNum\":4,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"BIOCHEMISTRY & MOLECULAR BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Receptors and Signal Transduction","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1080/10799893.2022.2116049","RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 1

摘要

E3泛素连接酶是细胞信号传导和蛋白平衡的重要调节因子,在包括癌症在内的许多疾病中受到严格控制。我们的研究旨在探讨E3泛素连接酶CBLC在乳腺癌中的生物学作用,并阐明CBLC介导的靶底物降解、细胞增殖和转移的特定机制网络。在这里,我们发现CBLC在乳腺癌组织和细胞中的表达高于正常组织和细胞。CBLC表达越高,乳腺癌患者预后越好。CBLC能抑制乳腺癌细胞的增殖、迁移和侵袭。Co-IP和免疫荧光共定位实验表明,CBLC与细胞质中的CTTN相互作用。CBLC通过泛素-蛋白酶体途径促进CTTN的降解,但不影响其mRNA水平。CBLC对乳腺癌细胞增殖、迁移和侵袭的抑制作用可被CTTN部分逆转。综上所述,我们的研究阐明了CBLC作为肿瘤抑制因子的生物学作用,发现了其功能底物,为CBLC/CTTN作为乳腺癌潜在治疗靶点提供了分子基础。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
CBLC inhibits the proliferation and metastasis of breast cancer cells via ubiquitination and degradation of CTTN.

The E3 ubiquitin ligase is an important regulator of cell signaling and proteostasis and is tightly controlled in many diseases, including cancer. Our study aimed to investigate the biological role of the E3 ubiquitin ligase CBLC in breast cancer and elucidate the specific mechanistic network underlying CBLC-mediated target substrate degradation, cell proliferation and metastasis. Here, we showed that CBLC expression was higher in breast cancer tissues and cells than that in normal tissues and cells. Higher expression of CBLC predicted a better prognosis for breast cancer patients. CBLC inhibited the proliferation, migration and invasion of breast cancer cells. Co-IP and immunofluorescence co-localization assays demonstrated that CBLC interacted with CTTN in the cytoplasm. CBLC promoted the degradation of CTTN through the ubiquitin-proteasome pathway without affecting its mRNA level. The inhibitory effect of CBLC on breast cancer cell proliferation, migration and invasion could partly be reversed by CTTN. Taken together, our study clarified the biological role of CBLC as a tumor suppressor and discovered its functional substrate, providing a molecular basis for CBLC/CTTN as a potential therapeutic target in breast cancer.

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
Journal of Receptors and Signal Transduction
Journal of Receptors and Signal Transduction 生物-生化与分子生物学
CiteScore
6.60
自引率
0.00%
发文量
19
审稿时长
>12 weeks
期刊介绍: Journal of Receptors and Signal Tranduction is included in the following abstracting and indexing services: BIOBASE; Biochemistry and Biophysics Citation Index; Biological Abstracts; BIOSIS Full Coverage Shared; BIOSIS Previews; Biotechnology Abstracts; Current Contents/Life Sciences; Derwent Chimera; Derwent Drug File; EMBASE; EMBIOLOGY; Journal Citation Reports/ Science Edition; PubMed/MedLine; Science Citation Index; SciSearch; SCOPUS; SIIC.
×
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