Polyoxometalate inhibition of SOX2-mediated tamoxifen resistance in breast cancer.

IF 8.2 2区 生物学 Q1 CELL BIOLOGY Cell Communication and Signaling Pub Date : 2024-09-02 DOI:10.1186/s12964-024-01800-w
Iskander Aurrekoetxea-Rodriguez, So Young Lee, Miriam Rábano, Isabel Gris-Cárdenas, Virginia Gamboa-Aldecoa, Irantzu Gorroño, Isabella Ramella-Gal, Connor Parry, Robert M Kypta, Beñat Artetxe, Juan M Gutierrez-Zorrilla, Maria dM Vivanco
{"title":"Polyoxometalate inhibition of SOX2-mediated tamoxifen resistance in breast cancer.","authors":"Iskander Aurrekoetxea-Rodriguez, So Young Lee, Miriam Rábano, Isabel Gris-Cárdenas, Virginia Gamboa-Aldecoa, Irantzu Gorroño, Isabella Ramella-Gal, Connor Parry, Robert M Kypta, Beñat Artetxe, Juan M Gutierrez-Zorrilla, Maria dM Vivanco","doi":"10.1186/s12964-024-01800-w","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Increased cancer stem cell (CSC) content and SOX2 overexpression are common features in the development of resistance to therapy in hormone-dependent breast cancer, which remains an important clinical challenge. SOX2 has potential as biomarker of resistance to treatment and as therapeutic target, but targeting transcription factors is also challenging. Here, we examine the potential inhibitory effect of different polyoxometalate (POM) derivatives on SOX2 transcription factor in tamoxifen-resistant breast cancer cells.</p><p><strong>Methods: </strong>Various POM derivatives were synthesised and characterised by infrared spectra, powder X-ray diffraction pattern and nuclear magnetic resonance spectroscopy. Estrogen receptor (ER) positive breast cancer cells, and their counterparts, which have developed resistance to the hormone therapy tamoxifen, were treated with POMs and their consequences assessed by gel retardation and chromatin immunoprecipitation to determine SOX2 binding to DNA. Effects on proliferation, migration, invasion and tumorigenicity were monitored and quantified using microscopy, clone formation, transwell, wound healing assays, flow cytometry and in vivo chick chorioallantoic membrane (CAM) models. Generation of lentiviral stable gene silencing and gene knock-out using CRISPR-Cas9 genome editing were applied to validate the inhibitory effects of the selected POM. Cancer stem cell subpopulations were quantified by mammosphere formation assays, ALDEFLUOR activity and CD44/CD24 stainings. Flow cytometry and western blotting were used to measure reactive oxygen species (ROS) and apoptosis.</p><p><strong>Results: </strong>POMs blocked in vitro binding activity of endogenous SOX2. [P<sub>2</sub>W<sub>18</sub>O<sub>62</sub>]<sup>6-</sup> (PW) Wells-Dawson-type anion was the most effective at inhibiting proliferation in various cell line models of tamoxifen resistance. 10 µM PW also reduced cancer cell migration and invasion, as well as SNAI2 expression levels. Treatment of tamoxifen-resistant cells with PW impaired tumour formation by reducing CSC content, in a SOX2-dependent manner, which led to stem cell depletion in vivo. Mechanistically, PW induced formation of reactive oxygen species (ROS) and inhibited Bcl-2, leading to the death of tamoxifen-resistant cells. PW-treated tamoxifen-resistant cells showed restored sensitivity to tamoxifen.</p><p><strong>Conclusions: </strong>Together, these observations highlight the potential use of PW as a SOX2 inhibitor and the therapeutic relevance of targeting SOX2 to treat tamoxifen-resistant breast cancer.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2000,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11367752/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Communication and Signaling","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s12964-024-01800-w","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Increased cancer stem cell (CSC) content and SOX2 overexpression are common features in the development of resistance to therapy in hormone-dependent breast cancer, which remains an important clinical challenge. SOX2 has potential as biomarker of resistance to treatment and as therapeutic target, but targeting transcription factors is also challenging. Here, we examine the potential inhibitory effect of different polyoxometalate (POM) derivatives on SOX2 transcription factor in tamoxifen-resistant breast cancer cells.

Methods: Various POM derivatives were synthesised and characterised by infrared spectra, powder X-ray diffraction pattern and nuclear magnetic resonance spectroscopy. Estrogen receptor (ER) positive breast cancer cells, and their counterparts, which have developed resistance to the hormone therapy tamoxifen, were treated with POMs and their consequences assessed by gel retardation and chromatin immunoprecipitation to determine SOX2 binding to DNA. Effects on proliferation, migration, invasion and tumorigenicity were monitored and quantified using microscopy, clone formation, transwell, wound healing assays, flow cytometry and in vivo chick chorioallantoic membrane (CAM) models. Generation of lentiviral stable gene silencing and gene knock-out using CRISPR-Cas9 genome editing were applied to validate the inhibitory effects of the selected POM. Cancer stem cell subpopulations were quantified by mammosphere formation assays, ALDEFLUOR activity and CD44/CD24 stainings. Flow cytometry and western blotting were used to measure reactive oxygen species (ROS) and apoptosis.

Results: POMs blocked in vitro binding activity of endogenous SOX2. [P2W18O62]6- (PW) Wells-Dawson-type anion was the most effective at inhibiting proliferation in various cell line models of tamoxifen resistance. 10 µM PW also reduced cancer cell migration and invasion, as well as SNAI2 expression levels. Treatment of tamoxifen-resistant cells with PW impaired tumour formation by reducing CSC content, in a SOX2-dependent manner, which led to stem cell depletion in vivo. Mechanistically, PW induced formation of reactive oxygen species (ROS) and inhibited Bcl-2, leading to the death of tamoxifen-resistant cells. PW-treated tamoxifen-resistant cells showed restored sensitivity to tamoxifen.

Conclusions: Together, these observations highlight the potential use of PW as a SOX2 inhibitor and the therapeutic relevance of targeting SOX2 to treat tamoxifen-resistant breast cancer.

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
多氧化金属盐抑制乳腺癌中 SOX2 介导的他莫昔芬抗药性
背景:癌症干细胞(CSC)含量增加和SOX2过表达是激素依赖性乳腺癌耐药性发展的共同特征,这仍是一项重要的临床挑战。SOX2具有作为耐药性生物标志物和治疗靶点的潜力,但靶向转录因子也具有挑战性。在此,我们研究了不同聚氧化金属(POM)衍生物对他莫昔芬耐药乳腺癌细胞中 SOX2 转录因子的潜在抑制作用:方法:合成了各种 POM 衍生物,并通过红外光谱、粉末 X 射线衍射图谱和核磁共振光谱对其进行了表征。用 POMs 处理雌激素受体(ER)阳性乳腺癌细胞及其对激素疗法他莫昔芬产生抗药性的对应细胞,并通过凝胶延缓和染色质免疫沉淀来评估其后果,以确定 SOX2 与 DNA 的结合情况。使用显微镜、克隆形成、跨孔、伤口愈合试验、流式细胞术和体内小鸡绒毛膜(CAM)模型监测和量化了对增殖、迁移、侵袭和致瘤性的影响。应用慢病毒稳定基因沉默和CRISPR-Cas9基因组编辑技术敲除基因,验证了所选POM的抑制作用。通过乳球形成试验、ALDEFLUOR活性和CD44/CD24染色对癌症干细胞亚群进行量化。流式细胞术和 Western 印迹法用于测量活性氧(ROS)和细胞凋亡:结果:POMs阻断了内源性SOX2的体外结合活性。[P2W18O62]6-(PW)威尔斯-道森型阴离子在各种他莫昔芬抗性细胞系模型中抑制增殖的效果最好。10 µM PW 还能降低癌细胞的迁移和侵袭以及 SNAI2 的表达水平。用PW处理他莫昔芬耐药细胞,能以SOX2依赖性方式减少癌细胞干细胞的含量,从而损害肿瘤的形成,导致体内干细胞耗竭。从机理上讲,PW诱导活性氧(ROS)的形成并抑制Bcl-2,导致他莫昔芬耐药细胞死亡。经PW处理的他莫昔芬耐药细胞恢复了对他莫昔芬的敏感性:这些观察结果凸显了PW作为SOX2抑制剂的潜在用途,以及靶向SOX2治疗他莫昔芬耐药乳腺癌的治疗意义。
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 去求助
来源期刊
CiteScore
11.00
自引率
0.00%
发文量
180
期刊介绍: Cell Communication and Signaling (CCS) is a peer-reviewed, open-access scientific journal that focuses on cellular signaling pathways in both normal and pathological conditions. It publishes original research, reviews, and commentaries, welcoming studies that utilize molecular, morphological, biochemical, structural, and cell biology approaches. CCS also encourages interdisciplinary work and innovative models, including in silico, in vitro, and in vivo approaches, to facilitate investigations of cell signaling pathways, networks, and behavior. Starting from January 2019, CCS is proud to announce its affiliation with the International Cell Death Society. The journal now encourages submissions covering all aspects of cell death, including apoptotic and non-apoptotic mechanisms, cell death in model systems, autophagy, clearance of dying cells, and the immunological and pathological consequences of dying cells in the tissue microenvironment.
期刊最新文献
ONC212, alone or in synergistic conjunction with Navitoclax (ABT-263), promotes cancer cell apoptosis via unconventional mitochondrial-independent caspase-3 activation Bivalent chromatin accommodates survivin and BRG1/SWI complex to activate DNA damage response in CD4+ cells Correction: Acidic preconditioning induced intracellular acid adaptation to protect renal injury via dynamic phosphorylation of focal adhesion kinase-dependent activation of sodium hydrogen exchanger 1 A novel amphiphilic squalene-based compound with open-chain polyethers reduces malignant melanoma metastasis in-vitro and in-vivo TMEM100 acts as a TAK1 receptor that prevents pathological cardiac hypertrophy progression
×
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