RBM7 deficiency promotes breast cancer metastasis by coordinating MFGE8 splicing switch and NF-kB pathway

Author:

Huang Fang1,Dai Zhenwei1,Yu Jinmiao1,Wang Kainan2,Chen Chaoqun1,Chen Dan3,Zhang Jinrui4,Zhao Jinyao4,Zhang Wenjing4,Qi Yangfan4,Wang Yang1ORCID

Affiliation:

1. Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University

2. Department of Oncology & Sino-US Research Center for Cancer Translational Medicine, the Second Affiliated Hospital, Dalian Medical University

3. Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University

4. Institute of Cancer Stem Cell, Dalian Medical University

Abstract

Aberrant alternative splicing is well-known to be closely associated with tumorigenesis of various cancers. However, the intricate mechanisms underlying breast cancer metastasis driven by deregulated splicing events remain largely unexplored. Here, we unveiled RBM7 as a novel regulator of alternative splicing that plays a crucial role in counteracting the metastatic potential of breast cancer. Through bioinformatics analysis and IHC staining validation, we revealed that RBM7 is decreased in lymph node and distant organ metastases of breast cancer as compared to primary lesions. Furthermore, we found that low expression of RBM7 is correlated with the reduced disease-free survival of breast cancer patients. Breast cancer cells with RBM7 depletion exhibited an increased potential for lung metastasis compared to scramble control cells. The absence of RBM7 stimulated breast cancer cell migration, invasion, and angiogenesis. Mechanistically, RBM7 regulates the splicing of MFGE8 directly, favoring the production of the predominant MFGE8-L isoform. This results in the attenuation of STAT1 phosphorylation and alterations in cell adhesion molecules. The MFGE8-L isoform exerted an inhibitory effect on the migratory and invasive capability of breast cancer cells, while the MFGE8-S isoform had the opposite effect. Particularly, the ectopic expression of MFGE8-L significantly reversed the pro-invasion effect of RBM7 silencing, but did not contribute to the promotion of angiogenesis-related secreted proteins. In RBM7 depleted cells, a gene set enrichment analysis revealed a significant amplification of the NF-κB cascade. Concordantly, RBM7 negatively regulates p65 phosphorylation. Furthermore, an NF-κB inhibitor could obstruct the increase in HUVEC tube formation caused by RBM7 silencing. Clinically, we noticed a positive correlation between RBM7 expression, MFGE-8 exon7 inclusion, and p65 downstream targets. Therefore, our study not only offer mechanistic insights into how abnormal splicing contributes to the aggressiveness of breast cancer, but also provide a new approach for molecular-targeted therapy in combating breast cancer.

Publisher

eLife Sciences Publications, Ltd

Reference39 articles.

1. SRSF1-Regulated Alternative Splicing in Breast Cancer;Molecular Cell,2015

2. Splicing in action: assessing disease causing sequence changes;Journal of Medical Genetics,2005

3. VEGF-A splicing: the key to anti-angiogenic therapeutics?;Bates SJHaDO;NATURE REVIEWS,2008

4. P-TEFb Activation by RBM7 Shapes a Pro-survival Transcriptional Response to Genotoxic Stress;Molecular Cell,2019

5. The biology and role of CD44 in cancer progression: therapeutic implications;Journal of Hematology & Oncology,2018

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3