Discover Oncology (Sep 2024)

Co-targeting NRF2 potentially enhances the in vitro anticancer effects of paclitaxel in gastric cancer cells

  • Shima Hasani,
  • Mohammad Khalaj-Kondori,
  • Sahar Safaei,
  • Mohammad Amini,
  • Negin Riazi-Tabrizi,
  • Mohadeseh Maghsoudi,
  • Behzad Baradaran

DOI
https://doi.org/10.1007/s12672-024-01312-6
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 14

Abstract

Read online

Abstract Background Gastric cancer (GC) is a highly chemoresistant malignancy with a poor prognosis. Paclitaxel’s low response rate as second-line chemotherapy for advanced GC has prompted intensive research into its molecular basis and prospective targeted therapies to enhance its therapeutic efficacy. The objective of this study was to investigate the synergistic effects of NRF2 silencing in combination with paclitaxel treatment on GC cell viability, apoptosis, proliferation, autophagy, and migration. Methods \After the siRNA-mediated silencing of NRF2 in AGS cells, the transfection efficacy was evaluated by qRT-PCR. The MTT assay was then applied to assess cell viability, followed by flow cytometry analysis for apoptosis, proliferation, and autophagy in AGS cells treated with NRF2 siRNA, paclitaxel, or their combination. Thereafter, the migration of cells was measured using a wound-healing assay. Ultimately, the relative gene expression levels of apoptotic (Bax, Caspase-3, and Caspase-9), anti-apoptotic (Bcl-2), metastatic (MMP-2), and cell cycle (P53) genes were measured by qRT-PCR in all experiment groups to further assess the molecular basis for the combination therapy. Results NRF2 siRNA transfection significantly enhanced paclitaxel-induced apoptosis and sensitized AGS cells to paclitaxel via modulating the expression of apoptosis-related genes including Bcl-2, Bax, Caspase-3, and Caspase-9. Besides, NRF2 siRNA and paclitaxel synergistically induced cell cycle arrest at the G2 phase, promoted autophagy activation, and inhibited AGS cell migration via MMP-2 downregulation. Additionally, P53, a key regulator of cell growth, was significantly upregulated in the treated groups compared to the control group. Conclusions Our findings suggest that paclitaxel combined with siRNA-mediated silencing of NRF2 might represent a promising therapeutic strategy for GC, however further translational and clinical research are warranted.

Keywords