Abstract

Simple SummaryCutaneous melanoma is the most aggressive form of skin cancer with high-metastatic ability. Despite the recent advancements in melanoma treatments, the prognosis of metastatic patients remains very poor. A better understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in order to develop novel therapeutical strategies to ameliorate patients’ outcomes. Extracellular vesicles (EV) released by tumor cells are key players in metastasis development: by conveying bioactive molecules with oncogenic activity, they can modulate the surrounding—and even the distant—microenvironment and reprogram recipient cells to facilitate the metastatic cascade. Here, we show that melanoma cells release a higher amount of miR-214-enriched EV when adapted to extracellular acidosis, which promote a macrophage activation state, capable of facilitating the trans-endothelial migration of melanoma cells. Thus, we disclose a new molecular mechanism to prevent melanoma intravasation based on miR-214 targeting.The understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in view of the identification of new targets and the development of innovative strategies to improve patients’ outcomes. Within the complexity of tumor intercellular communications leading to metastatic dissemination, extracellular vesicles (EV) released by tumor cells are central players. Indeed, the ability to travel through the circulatory system conveying oncogenic bioactive molecules even at distant sites makes EV capable of modulating recipient cells to facilitate metastatic dissemination. The dynamic remodeling of the tumor microenvironment might influence, along with a number of other events, tumoral EV release. We observed that, in melanoma, extracellular acidosis increases the release of EV enriched in miR-214, an onco-miRNA involved in melanoma metastasis. Then, miR-214-enriched EV were found to induce a state of macrophage activation, leading to an overproduction of proinflammatory cytokines and nitric oxide. Such an inflammatory microenvironment was able to alter the endothelial cell permeability, thereby facilitating the trans-endothelial migration of melanoma cells, a crucial step in the metastatic cascade. The use of synthetic miR-214 inhibitors and miR-214 overexpression allowed us to demonstrate the key role of miR-214 in the EV-dependent induction of macrophage activation. Overall, our in vitro study reveals that the release of tumor miR-214-enriched EV, potentiated by adapting tumor cells to extracellular acidosis, drives a macrophage-dependent trans-endothelial migration of melanoma cells. This finding points to miR-214 as a potential new therapeutic target to prevent melanoma intravasation.

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