SPI1-mediated transcriptional activation of CEP55 promotes the malignant growth of triple-negative breast cancer and M2 macrophage polarization

被引:1
|
作者
Liu, Yuanwei
Dong, Ming
Jia, Yong
Yang, Dezhen
Hui, Yang
Yang, Xiaodong [1 ]
机构
[1] Shaanxi Univ Chinese Med, Affiliated Hosp, Dept Surg Oncol, 2 Weiyang West Rd, Xianyang 712000, Peoples R China
关键词
TNBC; CEP55; SPI1; Macrophage polarization; MECHANISM;
D O I
10.1016/j.prp.2024.155544
中图分类号
R36 [病理学];
学科分类号
100104 ;
摘要
Background: Triple-negative breast cancer (TNBC) is a subtype of breast cancer that lacks the expression of three receptors commonly targeted in breast cancer treatment. In this study, the research focused on investigating the role of centrosomal protein 55 (CEP55) in TNBC progression and its interaction with the transcription factor Spi1 proto-oncogene (SPI1). Methods: Various techniques including qRT-PCR, western blotting, and immunohistochemistry assays were utilized to examine gene expression patterns. Functional assays such as wound-healing assay, transwell invasion assay, 5-Ethynyl-2'-deoxyuridine assay, and metabolic assays were conducted to assess the impact of CEP55 on the behaviors of TNBC cells. CD163-positive macrophages were quantified by flow cytometry. The chromatin immunoprecipitation assay and dual-luciferase reporter assay were performed to assess the association of SPI1 with CEP55. A xenograft mouse model experiment was used to analyze the impact of SPI1 on tumor development in vivo. Results: CEP55 and SPI1 expression levels were significantly upregulated in TNBC tissues and cells. The depletion of CEP55 led to decreased TNBC cell migration, invasion, proliferation, glucose metabolism, and M2 macrophage polarization, indicating its crucial role in promoting TNBC progression. Moreover, SPI1 transcriptionally activated CEP55 in TNBC cells, and its overexpression was associated with accelerated tumor growth in vivo. Further, CEP55 overexpression relieved SPI1 silencing-induced inhibitory effects on TNBC cell migration, invasion, proliferation, glucose metabolism, and M2 macrophage polarization. Conclusion: SPI1-mediated transcriptional activation of CEP55 plays a key role in enhancing TNBC cell migration, invasion, proliferation, glucose metabolism, and M2 macrophage polarization. These insights provide valuable information for potential targeted therapies to combat TNBC progression by modulating the SPI1-CEP55 axis.
引用
收藏
页数:9
相关论文
共 50 条
  • [31] A double feedback loop mediated by microRNA-23a/27a/24-2 regulates M1 versus M2 macrophage polarization and thus regulates cancer progression
    Ma, Sisi
    Liu, Min
    Xu, Zhenbiao
    Li, Yanshuang
    Guo, Hui
    Ge, Yehua
    Liu, Yanxin
    Zheng, Dexian
    Shi, Juan
    ONCOTARGET, 2016, 7 (12) : 13502 - 13519
  • [32] Circ_0062558 promotes growth, migration, and glutamine metabolism in triple-negative breast cancer by targeting the miR-876-3p/SLC1A5 axis
    Yuan, Mengzhen
    Zhang, Jun
    He, Yuxin
    Yi, Guangming
    Rong, Liwen
    Zheng, Liangjian
    Zhan, Tingting
    Zhou, Congming
    ARCHIVES OF GYNECOLOGY AND OBSTETRICS, 2022, 306 (05) : 1643 - 1655
  • [33] Circ_0062558 promotes growth, migration, and glutamine metabolism in triple-negative breast cancer by targeting the miR-876-3p/SLC1A5 axis
    Mengzhen Yuan
    Jun Zhang
    Yuxin He
    Guangming Yi
    Liwen Rong
    Liangjian Zheng
    Tingting Zhan
    Congming Zhou
    Archives of Gynecology and Obstetrics, 2022, 306 : 1643 - 1655
  • [34] PTTG1 promotes M2 macrophage polarization via the cGMP-PKG signaling pathway and facilitates EMT progression in human epithelial ovarian cancer cells
    Liang Tian
    Liyun Liu
    Chunlou Wang
    Yan Kong
    Zhigang Miao
    Qing Yao
    He Zhang
    Yuehong Li
    Discover Oncology, 16 (1)
  • [35] Upregulation of circ-UBAP2 predicts poor prognosis and promotes triple-negative breast cancer progression through the miR-661/MTA1 pathway
    Wang, Shengting
    Li, Qian
    Wang, Yufang
    Li, Xiaoming
    Wang, Rui
    Kang, Yuhua
    Xue, Xukai
    Meng, Rui
    Wei, Qi
    Feng, Xinghua
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, 2018, 505 (04) : 996 - 1002
  • [36] HTB50-2 Inhibits Growth and Migration of Triple-negative Breast Cancer via FOSL2/FOXC1 Signaling Axis and Subsequent Ferroptosis
    Liu, Na
    Yin, Wen-ying
    Duan, Wen-qi
    Lyu, Yu-zhu
    Xie, Shan
    Fang, Lei
    Ji, Yan
    Yang, Feng-ying
    Ge, Di
    CURRENT MEDICINAL CHEMISTRY, 2024,
  • [37] Tumor-derived exosomal miR-148b-3p mediates M2 macrophage polarization via TSC2/mTORC1 to promote breast cancer migration and invasion
    Hao, Chong
    Sheng, Zhimei
    Wang, Wenhao
    Feng, Ruijun
    Zheng, Yuanhang
    Xiao, Qinpei
    Zhang, Baogang
    THORACIC CANCER, 2023, 14 (16) : 1477 - 1491
  • [38] Long noncoding RNA-mediated activation of PROTOR1/PRR5-AKT signaling shunt downstream of PI3K in triple-negative breast cancer
    Tu, Zhenbo
    Hu, Yi
    Raizada, Devesh
    Bassal, Mahmoud A.
    Tenen, Daniel G.
    Karnoub, Antoine E.
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, 2022, 119 (43)
  • [39] Exosomal POU5 F1 derived from TNBC promotes cancer progression by regulating M2 macrophage polarization via inhibiting TRAF6 ubiquitination and activating AKT in macrophage
    Yimeng Chai
    Yao Shi
    Cell Biology and Toxicology, 41 (1)
  • [40] The RP11-417E7.1/THBS2 signaling pathway promotes colorectal cancer metastasis by activating the Wnt/β-catenin pathway and facilitating exosome-mediated M2 macrophage polarization
    Liu, Yunze
    Lv, Heng
    Liu, Xin
    Xu, Lei
    Li, Tiankang
    Zhou, Hui
    Zhu, Hongmei
    Hao, Chuanchuan
    Lin, Changwei
    Zhang, Yi
    JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH, 2024, 43 (01)