Your browser doesn't support javascript.
loading
Targeting IL-3Rα on tumor-derived endothelial cells blunts metastatic spread of triple-negative breast cancer via extracellular vesicle reprogramming.
Lopatina, Tatiana; Grange, Cristina; Cavallari, Claudia; Navarro-Tableros, Victor; Lombardo, Giusy; Rosso, Arturo; Cedrino, Massimo; Pomatto, Margherita Alba Carlotta; Koni, Malvina; Veneziano, Francesca; Castellano, Isabella; Camussi, Giovanni; Brizzi, Maria Felice.
Afiliação
  • Lopatina T; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Grange C; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Cavallari C; 2i3T Scarl University of Turin, Turin, Italy.
  • Navarro-Tableros V; 2i3T Scarl University of Turin, Turin, Italy.
  • Lombardo G; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Rosso A; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Cedrino M; 2i3T Scarl University of Turin, Turin, Italy.
  • Pomatto MAC; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Koni M; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Veneziano F; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Castellano I; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Camussi G; Department of Medical Sciences, University of Turin, Turin, Italy.
  • Brizzi MF; Department of Medical Sciences, University of Turin, Turin, Italy. mariafelice.brizzi@unito.it.
Oncogenesis ; 9(10): 90, 2020 Oct 10.
Article em En | MEDLINE | ID: mdl-33040091
ABSTRACT
The lack of approved targeted therapies highlights the need for new treatments for triple-negative breast cancer (TNBC) patients. Interleukin-3 (IL-3) acts as an autocrine factor for tumor-endothelial cells (TEC), and exerts pro-angiogenic paracrine action via extracellular vesicles (EVs). IL-3Rα blockade on TEC changes TEC-EV (anti-IL-3R-EV) microRNA (miR) content and promotes the regression of established vessels. As TEC is the doorway for "drug" entry into tumors, we aimed to assess whether IL-3R blockade on TEC impacts tumor progression via its unique EV cargo. First, the expression of IL-3Rα was evaluated in 27 human TNBC samples. It was noticed that, besides TEC and inflammatory cells, tumor cells from 55.5% of the human TNBC samples expressed IL-3Rα. Using human TNBC cell lines for in vitro studies, we found that, unlike native TEC-EVs (nEVs), anti-IL-3R-EVs increase apoptosis and reduced cell viability and migration. In vivo, anti-IL-3R-EV treatment induced vessel regression in established tumors formed of MDA-MB-231 cells, decreased Vimentin, ß-catenin, and TWIST1 expression, almost abolished liver and lung metastases from primary tumors, and reduced lung metastasis generated via the intravenous injection of MDA-MB-231 cells. nEVs depleted of miR-24-3p (antago-miR-24-3p-EVs) were effective as anti-IL-3R-EVs in downregulating TWIST1 and reducing metastatic lesions in vivo. Consistent with network analyses of miR-24-3p gene targeting, anti-IL-3R-EVs and antago-miR-24-3p-EVs upregulate SPRY2 in MDA-MB-231 cells. Finally, SPRY2 silencing prevented anti-IL-3R-EV and antago-miR-24-3p-EV-mediated apoptotic cues.Overall, these data provide the first evidence that IL-3Rα is highly expressed in TNBC cells, TEC, and inflammatory cells, and that IL-3Rα blockade on TEC impacts tumor progression.

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article