Abstract

Simple SummaryThe triple receptor negative breast cancer subtype, or TNBC, currently has no tailored treatment options. TNBC is highly metastatic, associated with high patient mortality, and disproportionately occurs in Black/African American women where it contributes to racial disparities in health outcomes. Therefore, we focused on new therapeutic approaches to TNBC. We discovered that levels of the Calcium-ATPase SPCA2 are abnormally low in TNBC and that these low levels correlate with poor survival prognosis in patients. Previously, we showed that recombinant SPCA2 prevented TNBC cells from acquiring aggressive “mesenchymal” properties associated with metastasis both in vitro and in vivo. These findings motivated us to search for drugs that turn the SPCA2 gene back on in TNBC cells. In this study, we show that histone deacetylase inhibitors increase SPCA2 levels, activate Ca2+ signaling and convert cancer cells to a less aggressive “epithelial” state. These findings could lead to new treatment options for TNBC.The secretory pathway Ca2+-ATPase SPCA2 is a tumor suppressor in triple receptor negative breast cancer (TNBC), a highly aggressive molecular subtype that lacks tailored treatment options. Low expression of SPCA2 in TNBC confers poor survival prognosis in patients. Previous work has established that re-introducing SPCA2 to TNBC cells restores basal Ca2+ signaling, represses mesenchymal gene expression, mitigates tumor migration in vitro and metastasis in vivo. In this study, we examined the effect of histone deacetylase inhibitors (HDACi) in TNBC cell lines. We show that the pan-HDACi vorinostat and the class I HDACi romidepsin induce dose-dependent upregulation of SPCA2 transcript with concurrent downregulation of mesenchymal markers and tumor cell migration characteristic of epithelial phenotype. Silencing SPCA2 abolished the ability of HDACi to reverse epithelial to mesenchymal transition (EMT). Independent of ATPase activity, SPCA2 elevated resting Ca2+ levels to activate downstream components of non-canonical Wnt/Ca2+ signaling. HDACi treatment led to SPCA2-dependent phosphorylation of CAMKII and β-catenin, turning Wnt signaling off. We conclude that SPCA2 mediates the efficacy of HDACi in reversing EMT in TNBC by a novel mode of non-canonical Wnt/Ca2+ signaling. Our findings provide incentive for screening epigenetic modulators that exploit Ca2+ signaling pathways to reverse EMT in breast tumors.

Highlights

  • Triple receptor negative breast cancer (TNBC) is a molecular subtype characterized by the lack of estrogen receptor (ER), progesterone receptor (PR), and receptor for human epidermal growth factor-2 (HER2), making it unsuitable for endocrine or targeted antibody therapy [1]

  • In proof-of-principle experiments we showed that ectopic expression of SPCA2 in TNBC cell lines enhanced post-translational stability of E-cadherin, repressed mesenchymal gene expression and tumor cell migration in vitro and mitigated tumor metastasis in vivo [4]

  • We have previously shown that ectopic expression of SPCA2 in triplenegative breast cancer elevates baseline Ca2+ via store independent Ca2+ entry (SICE) leading to phosphorylation and nuclear exclusion of YAP and suppression of epithelial to mesenchymal transition (EMT) [4]

Read more

Summary

Introduction

Triple receptor negative breast cancer (TNBC) is a molecular subtype characterized by the lack of estrogen receptor (ER), progesterone receptor (PR), and receptor for human epidermal growth factor-2 (HER2), making it unsuitable for endocrine or targeted antibody therapy [1]. Loss of SPCA2 and basal Ca2+ signaling in TNBC cells was found to promote epithelial–mesenchymal transition, a hallmark of metastasis, during which cancer cells lose their polarity and cell–cell contacts, thereby acquiring the ability to migrate [4]. EMT is characterized by gain in expression of mesenchymal genes, including N-cadherin and zinc-finger transcription factors such as Snail, Slug, and Zeb that reprogram the cancer cell to acquire malignant invasive phenotypes [5]. These gene expression changes serve as convenient markers to track reversible EMT changes in breast cancer cells

Methods
Results
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