文献检索文档翻译深度研究
Suppr Zotero 插件Zotero 插件
邀请有礼套餐&价格历史记录

新学期,新优惠

限时优惠:9月1日-9月22日

30天高级会员仅需29元

1天体验卡首发特惠仅需5.99元

了解详情
不再提醒
插件&应用
Suppr Zotero 插件Zotero 插件浏览器插件Mac 客户端Windows 客户端微信小程序
高级版
套餐订阅购买积分包
AI 工具
文献检索文档翻译深度研究
关于我们
关于 Suppr公司介绍联系我们用户协议隐私条款
关注我们

Suppr 超能文献

核心技术专利:CN118964589B侵权必究
粤ICP备2023148730 号-1Suppr @ 2025

TEAD4 的新靶标 RBM8A 通过调节 IGF1R 和 IRS-2 促进乳腺癌进展。

RBM8A, a new target of TEAD4, promotes breast cancer progression by regulating IGF1R and IRS-2.

机构信息

Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China.

Biomedical Experimental Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.

出版信息

J Transl Med. 2024 Sep 4;22(1):823. doi: 10.1186/s12967-024-05639-0.


DOI:10.1186/s12967-024-05639-0
PMID:39232805
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC11373126/
Abstract

BACKGROUND: Breast cancer (BC) is the most common malignant tumor in women worldwide, and further elucidation of the molecular mechanisms involved in BC pathogenesis is essential to improve the prognosis of BC patients. RNA Binding Motif Protein 8 A (RBM8A), with high affinity to a myriad of RNA transcripts, has been shown to play a crucial role in genesis and progression of multiple cancers. We attempted to explore its functional significance and molecular mechanisms in BC. METHODS: Bioinformatics analysis was performed on publicly available BC datasets. qRT-PCR was used to determine the expression of RBM8A in BC tissues. MTT assay, clone formation assay and flow cytometry were employed to examine BC cell proliferation and apoptosis in vitro. RNA immunoprecipitation (RIP) and RIP-seq were used to investigate the binding of RBM8A/EIF4A3 to the mRNA of IGF1R/IRS-2. RBM8A and EIF4A3 interactions were determined by co-immunoprecipitation (Co-IP) and immunofluorescence. Chromatin immunoprecipitation (Ch-IP) and dual-luciferase reporter assay were carried out to investigate the transcriptional regulation of RBM8A by TEAD4. Xenograft model was used to explore the effects of RBM8A and TEAD4 on BC cell growth in vivo. RESULTS: In this study, we showed that RBM8A is abnormally highly expressed in BC and knockdown of RBM8A inhibits BC cell proliferation and induces apoptosis in vitro. EIF4A3, which phenocopy RBM8A in BC, forms a complex with RBM8A in BC. Moreover, EIF4A3 and RBM8A complex regulate the expression of IGF1R and IRS-2 to activate the PI3K/AKT signaling pathway, thereby promoting BC progression. In addition, we identified TEAD4 as a transcriptional activator of RBM8A by Ch-IP, dual luciferase reporter gene and a series of functional rescue assays. Furthermore, we demonstrated the in vivo pro-carcinogenic effects of TEAD4 and RBM8A by xenograft tumor experiments in nude mice. CONCLUSION: Collectively, these findings suggest that TEAD4 novel transcriptional target RBM8A interacts with EIF4A3 to increase IGF1R and IRS-2 expression and activate PI3K/AKT signaling pathway, thereby further promoting the malignant phenotype of BC cells.

摘要

背景:乳腺癌(BC)是全球女性最常见的恶性肿瘤,进一步阐明 BC 发病机制中的分子机制对于改善 BC 患者的预后至关重要。RNA 结合基序蛋白 8A(RBM8A)与大量 RNA 转录本具有高亲和力,已被证明在多种癌症的发生和进展中发挥关键作用。我们试图探讨其在 BC 中的功能意义和分子机制。

方法:对公开的 BC 数据集进行生物信息学分析。qRT-PCR 用于检测 BC 组织中 RBM8A 的表达。MTT 测定、克隆形成测定和流式细胞术用于体外检测 BC 细胞增殖和凋亡。RNA 免疫沉淀(RIP)和 RIP-seq 用于研究 RBM8A/EIF4A3 与 IGF1R/IRS-2 mRNA 的结合。通过共免疫沉淀(Co-IP)和免疫荧光检测 RBM8A 和 EIF4A3 的相互作用。染色质免疫沉淀(Ch-IP)和双荧光素酶报告基因检测用于研究 TEAD4 对 RBM8A 的转录调控。通过异种移植模型研究 RBM8A 和 TEAD4 对体内 BC 细胞生长的影响。

结果:在这项研究中,我们表明 RBM8A 在 BC 中异常高表达,并且敲低 RBM8A 可抑制 BC 细胞的增殖并诱导体外凋亡。EIF4A3 在 BC 中与 RBM8A 具有相似表型,可与 RBM8A 在 BC 中形成复合物。此外,EIF4A3 和 RBM8A 复合物调节 IGF1R 和 IRS-2 的表达,激活 PI3K/AKT 信号通路,从而促进 BC 的进展。此外,我们通过 Ch-IP、双荧光素酶报告基因和一系列功能挽救实验鉴定了 TEAD4 是 RBM8A 的转录激活子。此外,我们通过裸鼠异种移植肿瘤实验证明了 TEAD4 和 RBM8A 在体内的致癌作用。

结论:总之,这些发现表明,TEAD4 的新转录靶标 RBM8A 与 EIF4A3 相互作用,增加 IGF1R 和 IRS-2 的表达并激活 PI3K/AKT 信号通路,从而进一步促进 BC 细胞的恶性表型。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/8a421f5a8a17/12967_2024_5639_Fig6_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/9669b7d4aae2/12967_2024_5639_Fig1_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/978670810d6c/12967_2024_5639_Fig2_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/c6d29b79009a/12967_2024_5639_Fig7_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/1ee03f09a036/12967_2024_5639_Fig3_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/9a06e744c808/12967_2024_5639_Fig8_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/5163ad7e157f/12967_2024_5639_Fig4_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/0b70c7238086/12967_2024_5639_Fig5_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/8a421f5a8a17/12967_2024_5639_Fig6_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/9669b7d4aae2/12967_2024_5639_Fig1_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/978670810d6c/12967_2024_5639_Fig2_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/c6d29b79009a/12967_2024_5639_Fig7_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/1ee03f09a036/12967_2024_5639_Fig3_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/9a06e744c808/12967_2024_5639_Fig8_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/5163ad7e157f/12967_2024_5639_Fig4_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/0b70c7238086/12967_2024_5639_Fig5_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/d07c/11373126/8a421f5a8a17/12967_2024_5639_Fig6_HTML.jpg

相似文献

[1]
RBM8A, a new target of TEAD4, promotes breast cancer progression by regulating IGF1R and IRS-2.

J Transl Med. 2024-9-4

[2]
TEAD4 modulated LncRNA MNX1-AS1 contributes to gastric cancer progression partly through suppressing BTG2 and activating BCL2.

Mol Cancer. 2020-1-10

[3]
TEAD4 Activates PCSK9 to Promote Stomach Adenocarcinoma Cell Stemness through Fatty Acid Metabolism.

Digestion. 2024

[4]
Metformin targets a YAP1-TEAD4 complex via AMPKα to regulate CCNE1/2 in bladder cancer cells.

J Exp Clin Cancer Res. 2019-8-27

[5]
TEAD4 functions as a prognostic biomarker and triggers EMT via PI3K/AKT pathway in bladder cancer.

J Exp Clin Cancer Res. 2022-5-17

[6]
RBM8A Depletion Decreases the Cisplatin Resistance and Represses the Proliferation and Metastasis of Breast Cancer Cells via AKT/mTOR Pathway.

Breast J. 2022

[7]
The interaction of TEA domain transcription factor 4 (TEAD4) and Yes-associated protein 1 (YAP1) promoted the malignant process mediated by serum/glucocorticoid regulated kinase 1 (SGK1).

Bioengineered. 2021-12

[8]
Circ-UBR1 facilitates proliferation, metastasis, and inhibits apoptosis in breast cancer by regulating the miR-1299/CCND1 axis.

Life Sci. 2021-2-1

[9]
PKP2 induced by YAP/TEAD4 promotes malignant progression of gastric cancer.

Mol Carcinog. 2024-9

[10]
m A modification of lncRNA PCAT6 promotes bone metastasis in prostate cancer through IGF2BP2-mediated IGF1R mRNA stabilization.

Clin Transl Med. 2021-6

引用本文的文献

[1]
Integrating single-cell regulatory atlas and multi-omics data for differential treatment response and multimodal predictive modeling in CDK 4/6 inhibitor-treated breast cancer.

Front Oncol. 2025-7-17

[2]
RBM8A promotes gastric cancer progression by binding with UPF3B to induce BBC3 mRNA degradation.

Int J Mol Med. 2025-9

本文引用的文献

[1]
EIF4A3-mediated biogenesis of circSTX6 promotes bladder cancer metastasis and cisplatin resistance.

J Exp Clin Cancer Res. 2024-1-2

[2]
IGF2BP3-EGFR-AKT axis promotes breast cancer MDA-MB-231 cell growth.

Biochim Biophys Acta Mol Cell Res. 2023-12

[3]
Bulk and single-cell transcriptome profiling reveal extracellular matrix mechanical regulation of lipid metabolism reprograming through YAP/TEAD4/ACADL axis in hepatocellular carcinoma.

Int J Biol Sci. 2023

[4]
The deubiquitinating enzyme UCHL3 promotes anaplastic thyroid cancer progression and metastasis through Hippo signaling pathway.

Cell Death Differ. 2023-5

[5]
MAD2L1 is transcriptionally regulated by TEAD4 and promotes cell proliferation and migration in colorectal cancer.

Cancer Gene Ther. 2023-5

[6]
Diverse Roles of the Exon Junction Complex Factors in the Cell Cycle, Cancer, and Neurodevelopmental Disorders-Potential for Therapeutic Targeting.

Int J Mol Sci. 2022-9-8

[7]
RNA binding protein POP7 regulates ILF3 mRNA stability and expression to promote breast cancer progression.

Cancer Sci. 2022-11

[8]
The Physiological Roles of the Exon Junction Complex in Development and Diseases.

Cells. 2022-4-1

[9]
Pan-cancer analysis, cell and animal experiments revealing TEAD4 as a tumor promoter in ccRCC.

Life Sci. 2022-3-15

[10]
The RNA-binding protein GRSF1 promotes hepatocarcinogenesis via competitively binding to YY1 mRNA with miR-30e-5p.

J Exp Clin Cancer Res. 2022-1-8

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

推荐工具

医学文档翻译智能文献检索