D-arabinose induces cell cycle arrest by promoting autophagy via p38 MAPK signaling pathway in breast cancer

Sci Rep. 2024 May 16;14(1):11219. doi: 10.1038/s41598-024-61309-7.

Abstract

Breast cancer patients often have a poor prognosis largely due to lack of effective targeted therapy. It is now well established that monosaccharide enhances growth retardation and chemotherapy sensitivity in tumor cells. We investigated whether D-arabinose has capability to restrict the proliferation of tumor cells and its mechanism. Here, we report that D-arabinose induced cytotoxicity is modulated by autophagy and p38 MAPK signaling pathway in breast cancer cell lines. The proliferation of cells was evaluated by CCK-8 and Colony formation assay. The distribution of cells in cell cycle phases was analyzed by flow cytometry. Cell cycle, autophagy and MAPK signaling related proteins were detected by western blotting. Mouse xenograft model was used to evaluate the efficacy of D-arabinose in vivo. The proliferation of cells was dramatically inhibited by D-arabinose exposure in a dose-dependent manner, which was relevant to cell cycle arrest, as demonstrated by G2/M cell cycle restriction and ectopic expression of cell cycle related proteins. Mechanistically, we further identified that D-arabinose is positively associated with autophagy and the activation of the p38 MAPK signaling in breast cancer. In contrast, 3-Ma or SB203580, the inhibitor of autophagy or p38 MAPK, reversed the efficacy of D-arabinose. Additionally, D-arabinose in vivo treatment could significantly inhibit xenograft growth of breast cancer cells. Our findings were the first to reveal that D-arabinose triggered cell cycle arrest by inducing autophagy through the activation of p38 MAPK signaling pathway in breast cancer cells.

Keywords: d-Arabinose; Autophagy; Breast cancer; Cell cycle arrest; p38 MAPK signaling pathway.

MeSH terms

  • Animals
  • Arabinose* / pharmacology
  • Autophagy* / drug effects
  • Breast Neoplasms* / drug therapy
  • Breast Neoplasms* / metabolism
  • Breast Neoplasms* / pathology
  • Cell Cycle Checkpoints* / drug effects
  • Cell Line, Tumor
  • Cell Proliferation* / drug effects
  • Female
  • Humans
  • MAP Kinase Signaling System* / drug effects
  • Mice
  • Mice, Inbred BALB C
  • Mice, Nude
  • Xenograft Model Antitumor Assays
  • p38 Mitogen-Activated Protein Kinases* / metabolism