Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | Breast Cancer | Research

Hypoxia-induced miR-181a-5p up-regulation reduces epirubicin sensitivity in breast cancer cells through inhibiting EPDR1/TRPC1 to activate PI3K/AKT signaling pathway

Authors: Yunwei Zhang, Yunping Guan, Xinyu Zheng, Chenyang Li

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Breast carcinoma (BC) ranks as a predominant malignancy and constitutes the second principal cause of mortality among women globally. Epirubicin stands as the drug of choice for BC therapeutics. Nevertheless, the emergence of chemoresistance has significantly curtailed its therapeutic efficacy. The resistance mechanisms to Epirubicin remain not entirely elucidated, yet they are conjectured to stem from diminished tumor vascular perfusion and resultant hypoxia consequent to Epirubicin administration. In our investigation, we meticulously scrutinized the Gene Expression Omnibus database for EPDR1, a gene implicated in hypoxia and Epirubicin resistance in BC. Subsequently, we delineated the impact of EPDR1 on cellular proliferation, motility, invasive capabilities, and interstitial-related proteins in BC cells, employing methodologies such as the CCK-8 assay, Transwell assay, and western blot analysis. Our research further unveiled that hypoxia-induced miR-181a-5p orchestrates the regulation of BC cell duplication, migration, invasion, and interstitial-related protein expression via modulation of EPDR1. In addition, we identified TRPC1, a gene associated with EPDR1 expression in BC, and substantiated that EPDR1 influences BC cellular dynamics through TRPC1-mediated modulation of the PI3K/AKT signaling cascade. Our findings underscore the pivotal role of EPDR1 in the development of BC. EPDR1 was found to be expressed at subdued levels in BC tissues, Epirubicin-resistant BC cells, and hypoxic BC cells. The overexpression of EPDR1 curtailed BC cell proliferation, motility, invasiveness, and the expression of interstitial-related proteins. At a mechanistic level, the overexpression of hypoxia-induced miR-181a-5p was observed to inhibit the EPDR1/TRPC1 axis, thereby activating the PI3K/AKT signaling pathway and diminishing the sensitivity to Epirubicin in BC cells. In summation, our study demonstrates that the augmentation of hypoxia-induced miR-181a-5p diminishes Epirubicin sensitivity in BC cells by attenuating EPDR1/TRPC1 expression, thereby invigorating the PI3K/AKT signaling pathway. This exposition offers a theoretical foundation for the application of Epirubicin in BC therapy, marking a significant contribution to the existing body of oncological literature.
Literature
2.
go back to reference Trayes KP, Cokenakes SEH. Breast Cancer Treatment. Am Fam Physician. 2021;104 2:171–8. Trayes KP, Cokenakes SEH. Breast Cancer Treatment. Am Fam Physician. 2021;104 2:171–8.
10.
go back to reference Vitale DL, Caon I, Parnigoni A, Sevic I, Spinelli FM, Icardi A, et al. Initial identification of UDP-Glucose dehydrogenase as a prognostic marker in breast Cancer patients, which facilitates Epirubicin Resistance and regulates Hyaluronan Synthesis in MDA-MB-231 cells. Biomolecules. 2021;11(2). https://doi.org/10.3390/biom11020246 Vitale DL, Caon I, Parnigoni A, Sevic I, Spinelli FM, Icardi A, et al. Initial identification of UDP-Glucose dehydrogenase as a prognostic marker in breast Cancer patients, which facilitates Epirubicin Resistance and regulates Hyaluronan Synthesis in MDA-MB-231 cells. Biomolecules. 2021;11(2). https://​doi.​org/​10.​3390/​biom11020246
19.
22.
28.
go back to reference Liang X, Bai J, Chen B. Overexpression of EPDR1 has an antitumorigenic effect on breast cancer in vitro. Int J Clin Exp Pathol. 2020;13 10:2628–36. Liang X, Bai J, Chen B. Overexpression of EPDR1 has an antitumorigenic effect on breast cancer in vitro. Int J Clin Exp Pathol. 2020;13 10:2628–36.
Metadata
Title
Hypoxia-induced miR-181a-5p up-regulation reduces epirubicin sensitivity in breast cancer cells through inhibiting EPDR1/TRPC1 to activate PI3K/AKT signaling pathway
Authors
Yunwei Zhang
Yunping Guan
Xinyu Zheng
Chenyang Li
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-11906-6

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine