Abstract

Simple SummaryBreast cancer accounts as the most extended disease among women worldwide. Store-operated calcium entry (SOCE), a major mechanism that allows calcium entry from the extracellular region through the plasma membrane, is required for several physiological processes. In recent years, it has been revealed that several breast cancer types present dysregulated calcium homeostasis, which contribute to their malignancy. Here we show the role of two important regulators of SOCE, SARAF and EFHB, which are necessary for cell viability, proliferation, and migration in breast cancer and pre-neoplastic cells, respectively, thus suggesting that these regulators play a key function in breast cancer development and progression.Breast cancer is among the most common malignancies in women. From the molecular point of view, breast cancer can be grouped into different categories, including the luminal (estrogen receptor positive (ER+)) and triple negative subtypes, which show distinctive features and, thus, are sensitive to different therapies. Breast cancer cells are strongly dependent on Ca2+ influx. Store-operated Ca2+ entry (SOCE) has been found to support a variety of cancer hallmarks including cell viability, proliferation, migration, and metastasis. The Ca2+ channels of the Orai family and the endoplasmic reticulum Ca2+ sensor STIM1 are the essential components of SOCE, but the extent of Ca2+ influx is fine-tuned by several regulatory proteins, such as the STIM1 modulators SARAF and EFHB. Here, we show that the expression and/or function of SARAF and EFHB is altered in breast cancer cells and both proteins are required for cell proliferation, migration, and viability. EFHB expression is upregulated in luminal and triple negative breast cancer (TNBC) cells and is essential for full SOCE in these cells. SARAF expression was found to be similar in breast cancer and pre-neoplastic breast epithelial cells, and SARAF knockdown was found to result in enhanced SOCE in pre-neoplastic and TNBC cells. Interestingly, silencing SARAF expression in ER+ MCF7 cells led to attenuation of SOCE, thus suggesting a distinctive role for SARAF in this cell type. Finally, we used a combination of approaches to show that molecular knockdown of SARAF and EFHB significantly attenuates the ability of breast cancer cells to proliferate and migrate, as well as cell viability. In aggregate, SARAF and EFHB are required for the fine modulation of SOCE in breast cancer cells and play an important role in the maintenance of proliferation, migration, and viability in these cells.

Highlights

  • Store-operated Ca2+ entry (SOCE), a mechanism for Ca2+ influx regulated by the filling state of the intracellular Ca2+ stores, is involved in the activation of cellular functions ranging from secretion to gene transcription, and dysregulation of this pathway has been associated to different disorders, including cancer [1]

  • We have explored the effect of cell transfection with shEFHB, shSARAF, or both on cell viability by using the cell-permeant dye calcein and propidium iodide

  • Our results indicate that EFHB and SARAF are expressed in neoplastic and preneoplastic cells

Read more

Summary

Introduction

Store-operated Ca2+ entry (SOCE), a mechanism for Ca2+ influx regulated by the filling state of the intracellular Ca2+ stores, is involved in the activation of cellular functions ranging from secretion to gene transcription, and dysregulation of this pathway has been associated to different disorders, including cancer [1]. Cancer cells are characterized by shifting the cell cycle control towards enhanced proliferation while suppressing cell death [2]. The mechanisms underlying these changes involve up- or downregulation of a variety of Ca2+ channels and regulatory proteins, including those involved in SOCE. Breast cancer is among the most common cancer types in women. SOCE through (or facilitated by) these channels has been reported to be critical for the development of a variety of breast cancer hallmarks, including cell proliferation, migration, and metastasis [9,10,11,12]

Methods
Findings
Discussion
Conclusion
Full Text
Published version (Free)

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