Abstract

Simple SummaryEarlier we demonstrated that the plasma membrane Ca2+ pump PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells, however, the exact mechanism has not been fully understood. Here we demonstrate that PMCA4b acted through actin cytoskeleton remodeling in generating a low migratory melanoma cell phenotype resulting in increased cell–cell connections, lamellipodia and stress fiber formation. Both proper trafficking and calcium transporting activity of the pump were essential to complete these tasks indicating that controlling Ca2+ concentration levels at specific plasma membrane locations such as the cell front played a role. Our findings suggest that PMCA4b downregulation is likely one of the mechanisms that leads to the perturbed cancer cell cytoskeleton organization resulting in enhanced melanoma cell migration and metastasis.We demonstrated that the plasma membrane Ca2+ ATPase PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells. Actin dynamics are essential for cells to move, invade and metastasize, therefore, we hypothesized that PMCA4b affected cell migration through remodeling of the actin cytoskeleton. We found that expression of PMCA4b in A375 BRAF mutant melanoma cells induced a profound change in cell shape, cell culture morphology, and displayed a polarized migratory character. Along with these changes the cells became more rounded with increased cell–cell connections, lamellipodia and stress fiber formation. Silencing PMCA4b in MCF-7 breast cancer cells had a similar effect, resulting in a dramatic loss of stress fibers. In addition, the PMCA4b expressing A375 cells maintained front-to-rear Ca2+ concentration gradient with the actin severing protein cofilin localizing to the lamellipodia, and preserved the integrity of the actin cytoskeleton from a destructive Ca2+ overload. We showed that both PMCA4b activity and trafficking were essential for the observed morphology and motility changes. In conclusion, our data suggest that PMCA4b plays a critical role in adopting front-to-rear polarity in a normally spindle-shaped cell type through F-actin rearrangement resulting in a less aggressive melanoma cell phenotype.

Highlights

  • Melanoma is a form of skin cancer that originates from the neural crest derived melanocytes that produce melanin

  • We investigated the role of PMCA4b activity and trafficking in maintaining the integrity of the actin cytoskeleton and we studied the formation of cell–cell connections, lamellipodia and stress fibers in cells expressing wild type and mutant PMCA4b proteins

  • Since plasma membrane Ca2+-ATPase (PMCA) are considered as key regulators of cellular Ca2+ homeostasis, we hypothesized that the reduced migration and morphology changes observed along with the reduced metastatic activity of these melanoma cells are correlated with changes in actin dynamics

Read more

Summary

Introduction

Melanoma is a form of skin cancer that originates from the neural crest derived melanocytes that produce melanin. It is a highly invasive type of cancer that tends to metastasize and causes death. It has been widely acknowledged that changes in the actin cytoskeleton arrangement are essential for cells to migrate and metastasize [2]. Ca2+ is involved in actin rearrangement, focal adhesion turnover, invadopodia, and lamellipodia formation during cell migration [8,9]. Changes in the expression of any of these tools can result in altered Ca2+ homeostasis leading to uncontrolled cell proliferation and metastasis [10]

Objectives
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