Oncotarget

Research Papers:

Enhancement of 5-FU sensitivity by the proapoptotic rpL3 gene in p53 null colon cancer cells through combined polymer nanoparticles

Annapina Russo _, Sara Maiolino, Valentina Pagliara, Francesca Ungaro, Fabiana Tatangelo, Alessandra Leone, Giulia Scalia, Alfredo Budillon, Fabiana Quaglia and Giulia Russo

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:79670-79687. https://doi.org/10.18632/oncotarget.13216

Metrics: PDF 2290 views  |   HTML 3624 views  |   ?  


Abstract

Annapina Russo1, Sara Maiolino2, Valentina Pagliara1, Francesca Ungaro2, Fabiana Tatangelo3, Alessandra Leone3, Giulia Scalia4, Alfredo Budillon3, Fabiana Quaglia2,*, Giulia Russo1,*

1Laboratory of Biochemistry, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy

2Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy

3Istituto Nazionale Tumori “Fondazione Pascale”-IRCCS, 80131 Napoli, Italy

4CEINGE-Biotecnologie Avanzate, 80145 Napoli, Italy

*These authors contributed equally to this work

Correspondence to:

Annapina Russo, email: [email protected]

Keywords: 5-FU, p53, ribosomal protein rpL3, colon cancer, apoptosis

Received: July 29, 2016     Accepted: October 14, 2016     Published: November 08, 2016

ABSTRACT

Colon cancer is one of the leading causes of cancer-related death worldwide and the therapy with 5-fluorouracil (5-FU) is mainly limited due to resistance. Recently, we have demonstrated that nucleolar stress upon 5-FU treatment leads to the activation of ribosome-free rpL3 (L3) as proapoptotic factor. In this study, we analyzed L3 expression profile in colon cancer tissues and demonstrated that L3 mRNA amount decreased with malignant progression and the intensity of its expression was inversely related to tumor grade and Bcl-2/Bax ratio. With the aim to develop a combined therapy of 5-FU plus plasmid encoding L3 (pL3), we firstly assessed the potentiation of the cytotoxic effect of 5-FU on colon cancer cells by L3. Next, 10 μM 5-FU and 2 μg of pL3 were encapsulated in biocompatible nanoparticles (NPs) chemically conjugated with HA to achieve active tumor-targeting ability in CD44 overexpressing cancer cells. We showed the specific intracellular accumulation of NPs in cells and a sustained release for 5-FU and L3. Analysis of cytotoxicity and apoptotic induction potential of combined NPs clearly showed that the 5-FU plus L3 were more effective in inducing apoptosis than 5-FU or L3 alone. Furthermore, we show that the cancer-specific chemosensitizer effect of combined NPs may be dependent on L3 ability to affect 5-FU efflux by controlling P-gp (P-glycoprotein) expression. These results led us to propose a novel combined therapy with the use of 5-FU plus L3 in order to establish individualized therapy by examining L3 profiles in tumors to yield a better clinical outcomes.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 13216