claudin -4-粘附信号通过肝脏X受体β驱动乳腺癌代谢和进展。

Yuko Murakami-Nishimagi, Kotaro Sugimoto, Makoto Kobayashi, Kazunoshin Tachibana, Manabu Kojima, Maiko Okano, Yuko Hashimoto, Shigehira Saji, Tohru Ohtake, Hideki Chiba
{"title":"claudin -4-粘附信号通过肝脏X受体β驱动乳腺癌代谢和进展。","authors":"Yuko Murakami-Nishimagi,&nbsp;Kotaro Sugimoto,&nbsp;Makoto Kobayashi,&nbsp;Kazunoshin Tachibana,&nbsp;Manabu Kojima,&nbsp;Maiko Okano,&nbsp;Yuko Hashimoto,&nbsp;Shigehira Saji,&nbsp;Tohru Ohtake,&nbsp;Hideki Chiba","doi":"10.1186/s13058-023-01646-z","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Cell adhesion is indispensable for appropriate tissue architecture and function in multicellular organisms. Besides maintaining tissue integrity, cell adhesion molecules, including tight-junction proteins claudins (CLDNs), exhibit the signaling abilities to control a variety of physiological and pathological processes. However, it is still fragmentary how cell adhesion signaling accesses the nucleus and regulates gene expression.</p><p><strong>Methods: </strong>By generating a number of knockout and rescued human breast cell lines and comparing their phenotypes, we determined whether and how CLDN4 affected breast cancer progression in vitro and in vivo. We also identified by RNA sequencing downstream genes whose expression was altered by CLDN4-adhesion signaling. Additionally, we analyzed by RT-qPCR the CLDN4-regulating genes by using a series of knockout and add-back cell lines. Moreover, by immunohistochemistry and semi-quantification, we verified the clinicopathological significance of CLDN4 and the nuclear receptor LXRβ (liver X receptor β) expression in breast cancer tissues from 187 patients.</p><p><strong>Results: </strong>We uncovered that the CLDN4-adhesion signaling accelerated breast cancer metabolism and progression via LXRβ. The second extracellular domain and the carboxy-terminal Y197 of CLDN4 were required to activate Src-family kinases (SFKs) and the downstream AKT in breast cancer cells to promote their proliferation. Knockout and rescue experiments revealed that the CLDN4 signaling targets the AKT phosphorylation site S432 in LXRβ, leading to enhanced cell proliferation, migration, and tumor growth, as well as cholesterol homeostasis and fatty acid metabolism, in breast cancer cells. In addition, RT-qPCR analysis showed the CLDN4-regulated genes are classified into at least six groups according to distinct LXRβ- and LXRβS432-dependence. Furthermore, among triple-negative breast cancer subjects, the \"CLDN4-high/LXRβ-high\" and \"CLDN4-low and/or LXRβ-low\" groups appeared to exhibit poor outcomes and relatively favorable prognoses, respectively.</p><p><strong>Conclusions: </strong>The identification of this machinery highlights a link between cell adhesion and transcription factor signalings to promote metabolic and progressive processes of malignant tumors and possibly to coordinate diverse physiological and pathological events.</p>","PeriodicalId":9283,"journal":{"name":"Breast Cancer Research : BCR","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10105442/pdf/","citationCount":"2","resultStr":"{\"title\":\"Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β.\",\"authors\":\"Yuko Murakami-Nishimagi,&nbsp;Kotaro Sugimoto,&nbsp;Makoto Kobayashi,&nbsp;Kazunoshin Tachibana,&nbsp;Manabu Kojima,&nbsp;Maiko Okano,&nbsp;Yuko Hashimoto,&nbsp;Shigehira Saji,&nbsp;Tohru Ohtake,&nbsp;Hideki Chiba\",\"doi\":\"10.1186/s13058-023-01646-z\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Cell adhesion is indispensable for appropriate tissue architecture and function in multicellular organisms. Besides maintaining tissue integrity, cell adhesion molecules, including tight-junction proteins claudins (CLDNs), exhibit the signaling abilities to control a variety of physiological and pathological processes. However, it is still fragmentary how cell adhesion signaling accesses the nucleus and regulates gene expression.</p><p><strong>Methods: </strong>By generating a number of knockout and rescued human breast cell lines and comparing their phenotypes, we determined whether and how CLDN4 affected breast cancer progression in vitro and in vivo. We also identified by RNA sequencing downstream genes whose expression was altered by CLDN4-adhesion signaling. Additionally, we analyzed by RT-qPCR the CLDN4-regulating genes by using a series of knockout and add-back cell lines. Moreover, by immunohistochemistry and semi-quantification, we verified the clinicopathological significance of CLDN4 and the nuclear receptor LXRβ (liver X receptor β) expression in breast cancer tissues from 187 patients.</p><p><strong>Results: </strong>We uncovered that the CLDN4-adhesion signaling accelerated breast cancer metabolism and progression via LXRβ. The second extracellular domain and the carboxy-terminal Y197 of CLDN4 were required to activate Src-family kinases (SFKs) and the downstream AKT in breast cancer cells to promote their proliferation. Knockout and rescue experiments revealed that the CLDN4 signaling targets the AKT phosphorylation site S432 in LXRβ, leading to enhanced cell proliferation, migration, and tumor growth, as well as cholesterol homeostasis and fatty acid metabolism, in breast cancer cells. In addition, RT-qPCR analysis showed the CLDN4-regulated genes are classified into at least six groups according to distinct LXRβ- and LXRβS432-dependence. Furthermore, among triple-negative breast cancer subjects, the \\\"CLDN4-high/LXRβ-high\\\" and \\\"CLDN4-low and/or LXRβ-low\\\" groups appeared to exhibit poor outcomes and relatively favorable prognoses, respectively.</p><p><strong>Conclusions: </strong>The identification of this machinery highlights a link between cell adhesion and transcription factor signalings to promote metabolic and progressive processes of malignant tumors and possibly to coordinate diverse physiological and pathological events.</p>\",\"PeriodicalId\":9283,\"journal\":{\"name\":\"Breast Cancer Research : BCR\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2023-04-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10105442/pdf/\",\"citationCount\":\"2\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Breast Cancer Research : BCR\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1186/s13058-023-01646-z\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Breast Cancer Research : BCR","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13058-023-01646-z","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 2

摘要

背景:细胞粘附是多细胞生物组织结构和功能的重要组成部分。除了维持组织的完整性,细胞粘附分子,包括紧密连接蛋白(cldn),表现出控制各种生理和病理过程的信号能力。然而,细胞粘附信号如何进入细胞核并调控基因表达仍不完整。方法:通过产生一些敲除和挽救的人乳腺细胞系,并比较它们的表型,我们在体外和体内确定CLDN4是否以及如何影响乳腺癌的进展。我们还通过RNA测序确定了表达被cldn4粘附信号改变的下游基因。此外,我们利用一系列敲除和加回细胞系,通过RT-qPCR分析了cldn4调节基因。此外,我们通过免疫组织化学和半定量方法验证了CLDN4和核受体LXRβ(肝X受体β)在187例乳腺癌组织中的表达的临床病理意义。结果:我们发现cldn4粘附信号通过LXRβ加速乳腺癌代谢和进展。CLDN4的第二个胞外结构域和羧基末端Y197是激活乳腺癌细胞中src家族激酶(SFKs)和下游AKT以促进其增殖所必需的。敲除和挽救实验表明,CLDN4信号通路靶向LXRβ中AKT磷酸化位点S432,从而增强乳腺癌细胞的增殖、迁移和肿瘤生长,以及胆固醇稳态和脂肪酸代谢。此外,RT-qPCR分析显示,根据LXRβ-和LXRβ s432依赖性的不同,cldn4调节基因可分为至少6组。此外,在三阴性乳腺癌受试者中,“cldn4 -高/ lxr β-高”组和“cldn4 -低和/或lxr β-低”组分别表现出较差的结局和相对有利的预后。结论:这一机制的发现强调了细胞粘附和转录因子信号之间的联系,以促进恶性肿瘤的代谢和进展过程,并可能协调各种生理和病理事件。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β.

Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β.

Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β.

Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β.

Background: Cell adhesion is indispensable for appropriate tissue architecture and function in multicellular organisms. Besides maintaining tissue integrity, cell adhesion molecules, including tight-junction proteins claudins (CLDNs), exhibit the signaling abilities to control a variety of physiological and pathological processes. However, it is still fragmentary how cell adhesion signaling accesses the nucleus and regulates gene expression.

Methods: By generating a number of knockout and rescued human breast cell lines and comparing their phenotypes, we determined whether and how CLDN4 affected breast cancer progression in vitro and in vivo. We also identified by RNA sequencing downstream genes whose expression was altered by CLDN4-adhesion signaling. Additionally, we analyzed by RT-qPCR the CLDN4-regulating genes by using a series of knockout and add-back cell lines. Moreover, by immunohistochemistry and semi-quantification, we verified the clinicopathological significance of CLDN4 and the nuclear receptor LXRβ (liver X receptor β) expression in breast cancer tissues from 187 patients.

Results: We uncovered that the CLDN4-adhesion signaling accelerated breast cancer metabolism and progression via LXRβ. The second extracellular domain and the carboxy-terminal Y197 of CLDN4 were required to activate Src-family kinases (SFKs) and the downstream AKT in breast cancer cells to promote their proliferation. Knockout and rescue experiments revealed that the CLDN4 signaling targets the AKT phosphorylation site S432 in LXRβ, leading to enhanced cell proliferation, migration, and tumor growth, as well as cholesterol homeostasis and fatty acid metabolism, in breast cancer cells. In addition, RT-qPCR analysis showed the CLDN4-regulated genes are classified into at least six groups according to distinct LXRβ- and LXRβS432-dependence. Furthermore, among triple-negative breast cancer subjects, the "CLDN4-high/LXRβ-high" and "CLDN4-low and/or LXRβ-low" groups appeared to exhibit poor outcomes and relatively favorable prognoses, respectively.

Conclusions: The identification of this machinery highlights a link between cell adhesion and transcription factor signalings to promote metabolic and progressive processes of malignant tumors and possibly to coordinate diverse physiological and pathological events.

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