A novel role for an RCAN3-derived peptide as a tumor suppressor in breast cancer

作者:Martinez Hoyer Sergio; Sole Sanchez Sonia; Aguado Fernando; Martinez Martinez Sara; Serrano Candelas Eva; Luis Hernandez Jose; Iglesias Mar; Miguel Redondo Juan; Casanovas Oriol; Messeguer Ramon; Perez Riba Merce*
来源:Carcinogenesis, 2015, 36(7): 792-799.
DOI:10.1093/carcin/bgv056

摘要

The conserved CIC motif of RCAN proteins mediates their interaction with the phosphatase CN. Here, we show that RCAN3 and a peptide derived from the CIC motif of RCAN3 protein block tumor progression by inhibiting CN-NFATc signaling pathway.The members of the human regulators of calcineurin (RCAN) protein family are endogenous regulators of the calcineurin (CN)-cytosolic nuclear factor of activated T-cells (NFATc) pathway activation. This function is explained by the presence of a highly conserved calcipressin inhibitor of calcineurin (CIC) motif in RCAN proteins, which has been shown to compete with NFATc for the binding to CN and therefore are able to inhibit NFATc dephosphorylation and activation by CN. Very recently, emerging roles for NFATc proteins in transformation, tumor angiogenesis and metastasis have been described in different cancer cell types. In this work, we report that the overexpression of RCAN3 dramatically inhibits tumor growth and tumor angiogenesis in an orthotopic human breast cancer model. We suggest that RCAN3 exerts these effects in a CN-dependent manner, as mutation of the CIC motif in RCAN3 abolishes the tumor suppressor effect. Moreover, the expression of the EGFP-R3(178-210) peptide, spanning the CIC motif of RCAN3, is able to reproduce all the antitumor effects of RCAN3 full-length protein. Finally, we show that RCAN3 and the EGFP-R3(178-210) peptide inhibit the CN-NFATc signaling pathway and the induction of the NFATc-dependent gene cyclooxygenase-2. Our work suggests that the EGFP-R3(178-210) peptide possess potent tumor suppressor properties and therefore constitutes a novel lead for the development of potent and specific antitumoral agents. Moreover, we propose the targeting of the CN-NFATc pathway in the tumor cells constitutes an effective way to hamper tumor progression by impairing the paracrine network among tumor, endothelial and polymorphonucleated cells.

  • 出版日期2015-7