Abstract

Tumor cell heterogeneity is primarily dictated by mutational changes, sometimes leading to clones that undergo a metastatic switch. However, little is known about tumor heterogeneity following chemotherapy perturbation. Here we studied the possible involvement of tumor-derived extracellular vesicles, often referred to as tumor-derived microparticles (TMPs), as mediators of the metastatic switch in the tumor microenvironment by hindering cell adhesion properties. Specifically, we show that highly metastatic or chemotherapy-treated breast cancer cells shed an increased number of TMPs compared to their respective controls. We found that these TMPs substantially reduce cell adhesion and disrupt actin filament structure, therefore increasing their biomechanical force pace, further implicating tumor cell dissemination as part of the metastatic cascade. Our results demonstrate that these pro-metastatic effects are mediated in part by CD44 which is highly expressed in TMPs obtained from highly metastatic cells or cells exposed to chemotherapy when compared to cells with low metastatic potential. Consequently, when we inhibited CD44 expression on TMPs by a pharmacological or a genetic approach, increased tumor cell adhesion and re-organized actin filament structure were observed. We also demonstrated that breast cancer patients treated with paclitaxel chemotherapy exhibited increased CD44-expressing TMPs. Overall, our study provides further insights into the role of TMPs in promoting metastasis, an effect which is augmented when tumor cells are exposed to chemotherapy.

Highlights

  • Significant progress has been made in the last decades towards the development of novel anti-cancer therapies for the treatment of advanced metastatic disease, most cancer types areCells 2020, 9, 2269; doi:10.3390/cells9102269 www.mdpi.com/journal/cellsCells 2020, 9, 2269 still incurable, with metastasis being the main cause of death

  • The number of tumor-derived microparticles (TMPs) extracted from highly metastatic cells or from cells exposed to chemotherapy was higher than control cells, as previously reported [10,14], their size was not significantly different as evaluated by NanoSight (Figure S1)

  • We postulated that TMPs from highly metastatic cells or from cells exposed to chemotherapy could augment the metastatic potential of low-metastatic cells

Read more

Summary

Introduction

Significant progress has been made in the last decades towards the development of novel anti-cancer therapies for the treatment of advanced metastatic disease, most cancer types areCells 2020, 9, 2269; doi:10.3390/cells9102269 www.mdpi.com/journal/cellsCells 2020, 9, 2269 still incurable, with metastasis being the main cause of death. Metastasis is a multi-step process that includes cancer cell dissemination from the primary tumor, intravasation to the blood or lymphatic system, survival in the circulation, extravasation to a target organ, and seeding and proliferation at a distant site [3,4]. These effects require a tight regulation of the cellular machinery that supports tumor cell detachment from the primary tumor and their binding to the metastatic site. At remote secondary sites, the EVs are taken up by organ-derived cells, thereby preparing the microenvironment for future tumor cell seeding. EVs play a significant role in tumor growth and metastasis

Methods
Results
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.