Abstract

BackgroundUnlike other breast cancer subtypes that may be treated with a variety of hormonal or targeted therapies, there is a need to identify new, effective targets for triple-negative breast cancer (TNBC). It has recently been recognized that membrane potential is depolarized in breast cancer cells. The primary objective of the study is to explore whether hyperpolarization induced by opening potassium channels may provide a new strategy for treatment of TNBC.MethodsBreast cancer datasets in cBioPortal for cancer genomics was used to search for ion channel gene expression. Immunoblots and immunohistochemistry were used for protein expression in culture cells and in the patient tissues. Electrophysiological patch clamp techniques were used to study properties of BK channels in culture cells. Flow cytometry and fluorescence microscope were used for cell viability and cell cycle studies. Ultrasound imaging was used to study xenograft in female NSG mice.ResultsIn large datasets of breast cancer patients, we identified a gene, KCNMA1 (encoding for a voltage- and calcium-dependent large-conductance potassium channel, called BK channel), overexpressed in triple-negative breast cancer patients. Although overexpressed, 99% of channels are closed in TNBC cells. Opening BK channels hyperpolarized membrane potential, which induced cell cycle arrest in G2 phase and apoptosis via caspase-3 activation. In a TNBC cell induced xenograft model, treatment with a BK channel opener significantly slowed tumor growth without cardiac toxicity.ConclusionsOur results support the idea that hyperpolarization induced by opening BK channel in TNBC cells can become a new strategy for development of a targeted therapy in TNBC.

Highlights

  • Unlike other breast cancer subtypes that may be treated with a variety of hormonal or targeted therapies, there is a need to identify new, effective targets for triple-negative breast cancer (TNBC)

  • Previous studies have demonstrated that Big K (BK) channel alpha subunit protein is abundantly expressed in MDA-Mouse brain (MB)-231 cells, weakly expressed in MCF7, and nearly undetectable in normal breast epithelial cells MCF10A [15]

  • Voltage-dependent activation of BK channels in TNBC cells Previously, we showed that the resting Em, which is within the physiological voltage range, in MDA-MB-231 cells is depolarized compared to normal mammary epithelial cells (HMEC) (Em_MDA-MB-231: about -40 mV, Em_HMEC: about -67 mV) [12]

Read more

Summary

Introduction

Unlike other breast cancer subtypes that may be treated with a variety of hormonal or targeted therapies, there is a need to identify new, effective targets for triple-negative breast cancer (TNBC). Further studies on molecular signatures, genetics, and genomics have led to the identification of four TNBC subtypes (basallike 1, basal-like 2, mesenchymal, and luminal androgen receptor) [4, 5]. These studies have revealed the complexity of breast tumors and generated many new hypotheses for potential therapeutic targets for treatment of TNBC. There is a need to identify new, effective targets for TNBC

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.