Abstract

Breast cancer MCF-7 cell-line-derived mammospheres were shown to be enriched in cells with a CD44+/CD24– surface profile, consistent with breast cancer stem cells (BCSC). These BCSC were previously reported to express key sphingolipid signaling effectors, including pro-oncogenic sphingosine kinase 1 (SphK1) and sphingosine-1-phosphate receptor 3 (S1P3). In this study, we explored intracellular trafficking and localization of SphK1 and S1P3 in parental MCF-7 cells, and MCF-7 derived BCSC-enriched mammospheres treated with growth- or apoptosis-stimulating agents. Intracellular trafficking and localization were assessed using confocal microscopy and cell fractionation, while CD44+/CD24- marker status was confirmed by flow cytometry. Mammospheres expressed significantly higher levels of S1P3 compared to parental MCF-7 cells (p < 0.01). Growth-promoting agents (S1P and estrogen) induced SphK1 and S1P3 translocation from cytoplasm to nuclei, which may facilitate the involvement of SphK1 and S1P3 in gene regulation. In contrast, pro-apoptotic cytokine tumor necrosis factor α (TNFα)-treated MCF-7 cells demonstrated increased apoptosis and no nuclear localization of SphK1 and S1P3, suggesting that TNFα can inhibit nuclear translocation of SphK1 and S1P3. TNFα inhibited mammosphere formation and induced S1P3 internalization and degradation. No nuclear translocation of S1P3 was detected in TNFα-stimulated mammospheres. Notably, SphK1 and S1P3 expression and localization were highly heterogenous in mammospheres, suggesting the potential for a large variety of responses. The findings provide further insights into the understanding of sphingolipid signaling and intracellular trafficking in BCs. Our data indicates that the inhibition of SphK1 and S1P3 nuclear translocation represents a novel method to prevent BCSCs proliferation.

Highlights

  • Sphingolipids, their receptors, and sphingolipid-metabolizing enzymes support and regulate the growth and survival of both normal and malignant cells

  • Considering that compartmentalization of the protein defines the range of its potential signaling partners and mechanism of action, the purpose of this study was to visualize the localization of sphingosine kinase 1 (SphK1) and sphingosine-1-phosphate receptor 3 (S1P3) in cells treated by SphK1- and S1P3-activating substances with growth- or apoptosis-promoting effects

  • The findings indicate that S1P and estrogen stimulate translocation of SphK1 and S1P3 into the MCF-7 cell nuclei

Read more

Summary

Introduction

Sphingolipids, their receptors, and sphingolipid-metabolizing enzymes support and regulate the growth and survival of both normal and malignant cells. Sphingolipids operate as signaling molecules and control numerous biological processes including cell division, differentiation, migration, and survival; they have been associated with malignant transformation [1,2,3]. Sphingolipid-modifying enzymes and receptors represent attractive therapeutic targets for a range of diseases, including cancer. Blocking and/or modifying agents that regulate sphingosine kinases (SphK1 and SphK2) and sphingosine-1-phosphate (S1P) receptor-signaling pathways have been intensively explored in breast cancer [3,4,5]. SphK2 was shown to transmit antiproliferative signals and its nuclear presence is well documented [6]. This study investigated growth- or apoptosis-triggered SphK1 trafficking in breast cancer cells

Objectives
Methods
Results
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