1. Academic Validation
  2. uPAR Controls Vasculogenic Mimicry Ability Expressed by Drug-Resistant Melanoma Cells

uPAR Controls Vasculogenic Mimicry Ability Expressed by Drug-Resistant Melanoma Cells

  • Oncol Res. 2022 Jan 31;28(9):873-884. doi: 10.3727/096504021X16273798026651.
Elena Andreucci 1 Anna Laurenzana 1 Silvia Peppicelli 1 Alessio Biagioni 1 Francesca Margheri 1 Jessica Ruzzolini 1 Francesca Bianchini 1 Gabriella Fibbi 1 Mario Del Rosso 1 Chiara Nediani 1 Simona Serrat 2 Livia Fucci 3 Michele Guida 4 Lido Calorini 1
Affiliations

Affiliations

  • 1 Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of FlorenceFlorenceItaly.
  • 2 Laboratory of Nanotecnology, IRCCS Istituto Tumori Giovanni Paolo II,BariItaly.
  • 3 Pathology Department, IRCCS Istituto Tumori Giovanni Paolo II,BariItaly.
  • 4 Rare Tumors and Melnaoma Unit, IRCCS Istituto Tumori Giovanni Paolo II,BariItaly.
Abstract

Malignant melanoma is a highly aggressive skin Cancer characterized by an elevated grade of tumor cell plasticity. Such plasticity allows adaptation of melanoma cells to different hostile conditions and guarantees tumor survival and disease progression, including aggressive features such as drug resistance. Indeed, almost 50% of melanoma rapidly develop resistance to the BRafV600E inhibitor vemurafenib, with fast tumor dissemination, a devastating consequence for patients outcomes. Vasculogenic mimicry (VM), the ability of Cancer cells to organize themselves in perfused vascular-like channels, might sustain tumor spread by providing vemurafenib-resistant Cancer cells with supplementary ways to enter into circulation and disseminate. Thus, this research aims to determine if vemurafenib resistance goes with the acquisition of VM ability by aggressive melanoma cells, and identify a driving molecule for both vemurafenib resistance and VM. We used two independent experimental models of drug-resistant melanoma cells, the first one represented by a chronic adaptation of melanoma cells to extracellular acidosis, known to drive a particularly aggressive and vemurafenib-resistant phenotype, the second one generated with chronic vemurafenib exposure. By performing in vitro tube formation assay and evaluating the expression levels of the VM markers EphA2 and VE-cadherin by Western blotting and flow cytometer analyses, we demonstrated that vemurafenib-resistant cells obtained by both models are characterized by an increased ability to perform VM. Moreover, by exploiting the CRISPR-Cas9 technique and using the urokinase plasminogen activator receptor (uPAR) inhibitor M25, we identified uPAR as a driver of VM expressed by vemurafenib-resistant melanoma cells. Thus, uPAR targeting may be successfully leveraged as a new complementary therapy to inhibit VM in drug-resistant melanoma patients, to counteract the rapid progression and dissemination of the disease.

Figures
Products