Enhanced Intracellular Reactive Oxygen Species by Photodynamic Therapy Effectively Promotes Chemoresistant Cell Death

Int J Biol Sci. 2022 Jan 1;18(1):374-385. doi: 10.7150/ijbs.66602. eCollection 2022.

Abstract

Anti-cancer chemo-drugs can cause a rapid elevation of intracellular reactive oxygen species (ROS) levels. An imbalance in ROS production and elimination systems leads to cancer cell resistance to chemotherapy. This study aimed to evaluate the mechanism and effect of ROS on multidrug resistance in various human chemoresistant cancer cells by detecting the changes in the amount of ROS, the expression of ROS-related and glycolysis-related genes, and cell death. We found that ROS was decreased while oxidative phosphorylation was increased in chemoresistant cells. We verified that the chemoresistance of cancer cells was achieved in two ways. First, chemoresistant cells preferred oxidative phosphorylation instead of anaerobic glycolysis for energy generation, which increased ATPase activity and produced much more ATP to provide energy. Second, ROS-scavenging systems were enhanced in chemoresistant cancer cells, which in turn decreased ROS amount and thus inhibited chemo-induced cell death. Our in vitro and in vivo photodynamic therapy further demonstrated that elevated ROS production efficiently inhibited chemo-drug resistance and promoted chemoresistant cell death. Taken together, targeting ROS systems has a great potential to treat cancer patients with chemoresistance.

Keywords: Chemoresistance; ROS; glycolysis; malignant tumor; oxidative phosphorylation; photodynamic therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Death*
  • Cell Line, Tumor
  • Drug Resistance, Neoplasm / drug effects*
  • Female
  • Glycolysis
  • Humans
  • Mice
  • Mice, Inbred BALB C
  • Oxidative Phosphorylation*
  • Photochemotherapy / methods*
  • Reactive Oxygen Species / metabolism*
  • Xenograft Model Antitumor Assays

Substances

  • Reactive Oxygen Species