Cooperative NF-κB and Notch1 signaling promotes macrophage-mediated MenaINV expression in breast cancer.

Camille L Duran, George S Karagiannis, Xiaoming Chen, Ved P Sharma, David Entenberg, John S Condeelis, Maja H Oktay
{"title":"Cooperative NF-κB and Notch1 signaling promotes macrophage-mediated MenaINV expression in breast cancer.","authors":"Camille L Duran, George S Karagiannis, Xiaoming Chen, Ved P Sharma, David Entenberg, John S Condeelis, Maja H Oktay","doi":"10.1186/s13058-023-01628-1","DOIUrl":null,"url":null,"abstract":"<p><p>Metastasis is a multistep process that leads to the formation of clinically detectable tumor foci at distant organs and frequently to patient demise. Only a subpopulation of breast cancer cells within the primary tumor can disseminate systemically and cause metastasis. To disseminate, cancer cells must express MenaINV, an isoform of the actin regulatory protein Mena, encoded by the ENAH gene, that endows tumor cells with transendothelial migration activity, allowing them to enter and exit the blood circulation. We have previously demonstrated that MenaINV mRNA and protein expression is induced in cancer cells by macrophage contact. In this study, we discovered the precise mechanism by which macrophages induce MenaINV expression in tumor cells. We examined the promoter of the human and mouse ENAH gene and discovered a conserved NF-κB transcription factor binding site. Using live imaging of an NF-κB activity reporter and staining of fixed tissues from mouse and human breast cancer, we further determined that for maximal induction of MenaINV in cancer cells, NF-κB needs to cooperate with the Notch1 signaling pathway. Mechanistically, Notch1 signaling does not directly increase MenaINV expression, but it enhances and sustains NF-κB signaling through retention of p65, an NF-κB transcription factor, in the nucleus of tumor cells, leading to increased MenaINV expression. In mice, these signals are augmented following chemotherapy treatment and abrogated upon macrophage depletion. Targeting Notch1 signaling in vivo decreased NF-κB signaling activation and MenaINV expression in the primary tumor and decreased metastasis. Altogether, these data uncover mechanistic targets for blocking MenaINV induction that should be explored clinically to decrease cancer cell dissemination and improve survival of patients with metastatic disease.</p>","PeriodicalId":9283,"journal":{"name":"Breast Cancer Research : BCR","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-04-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10080980/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Breast Cancer Research : BCR","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13058-023-01628-1","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Metastasis is a multistep process that leads to the formation of clinically detectable tumor foci at distant organs and frequently to patient demise. Only a subpopulation of breast cancer cells within the primary tumor can disseminate systemically and cause metastasis. To disseminate, cancer cells must express MenaINV, an isoform of the actin regulatory protein Mena, encoded by the ENAH gene, that endows tumor cells with transendothelial migration activity, allowing them to enter and exit the blood circulation. We have previously demonstrated that MenaINV mRNA and protein expression is induced in cancer cells by macrophage contact. In this study, we discovered the precise mechanism by which macrophages induce MenaINV expression in tumor cells. We examined the promoter of the human and mouse ENAH gene and discovered a conserved NF-κB transcription factor binding site. Using live imaging of an NF-κB activity reporter and staining of fixed tissues from mouse and human breast cancer, we further determined that for maximal induction of MenaINV in cancer cells, NF-κB needs to cooperate with the Notch1 signaling pathway. Mechanistically, Notch1 signaling does not directly increase MenaINV expression, but it enhances and sustains NF-κB signaling through retention of p65, an NF-κB transcription factor, in the nucleus of tumor cells, leading to increased MenaINV expression. In mice, these signals are augmented following chemotherapy treatment and abrogated upon macrophage depletion. Targeting Notch1 signaling in vivo decreased NF-κB signaling activation and MenaINV expression in the primary tumor and decreased metastasis. Altogether, these data uncover mechanistic targets for blocking MenaINV induction that should be explored clinically to decrease cancer cell dissemination and improve survival of patients with metastatic disease.

Abstract Image

Abstract Image

Abstract Image

NF-κB和Notch1信号的协同作用促进了巨噬细胞介导的MenaINV在乳腺癌中的表达。
转移是一个多步骤的过程,它导致在远处器官形成临床上可检测到的肿瘤病灶,并常常导致患者死亡。只有原发肿瘤内的乳腺癌细胞亚群才能向全身扩散并导致转移。要进行扩散,癌细胞必须表达 MenaINV,这是一种由ENAH基因编码的肌动蛋白调节蛋白Mena的异构体,它赋予肿瘤细胞跨内皮迁移活性,使其能够进出血液循环。我们以前曾证实,巨噬细胞接触会诱导癌细胞中 MenaINV mRNA 和蛋白的表达。在这项研究中,我们发现了巨噬细胞在肿瘤细胞中诱导 MenaINV 表达的确切机制。我们研究了人类和小鼠ENAH基因的启动子,发现了一个保守的NF-κB转录因子结合位点。通过对NF-κB活性报告因子的实时成像以及对小鼠和人类乳腺癌固定组织的染色,我们进一步确定,要在癌细胞中最大程度地诱导MenaINV,NF-κB需要与Notch1信号通路合作。从机制上讲,Notch1 信号并不直接增加 MenaINV 的表达,但它通过在肿瘤细胞核中保留 NF-κB 转录因子 p65 来增强和维持 NF-κB 信号,从而导致 MenaINV 表达的增加。在小鼠体内,这些信号在化疗后会增强,而在巨噬细胞耗竭后会减弱。在体内靶向 Notch1 信号可减少 NF-κB 信号的激活和原发肿瘤中 MenaINV 的表达,并减少转移。总之,这些数据揭示了阻断MenaINV诱导的机理靶点,应在临床上加以探索,以减少癌细胞扩散,提高转移性疾病患者的生存率。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
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