1. Academic Validation
  2. Chlorin e6-Induced Photodynamic Effect Polarizes the Macrophage Into an M1 Phenotype Through Oxidative DNA Damage and Activation of STING

Chlorin e6-Induced Photodynamic Effect Polarizes the Macrophage Into an M1 Phenotype Through Oxidative DNA Damage and Activation of STING

  • Front Pharmacol. 2022 Mar 3;13:837784. doi: 10.3389/fphar.2022.837784.
Ting-Ting Yu 1 2 3 Ning Han 1 2 3 Liu-Gen Li 1 3 Xing-Chun Peng 1 3 Qi-Rui Li 1 3 Hua-Zhen Xu 4 Xi-Yong Wang 1 3 Zi-Yi Yang 1 3 Xiao Chen 4 Mei-Fang Wang 2 3 Tong-Fei Li 1 2 3
Affiliations

Affiliations

  • 1 School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.
  • 2 Department of Respiratory, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China.
  • 3 Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China.
  • 4 Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
Abstract

The tumor-associated macrophage (TAM) serves as an immunosuppressive agent in the malignant tumor microenvironment, facilitating the development and metastasis of lung Cancer. The photodynamic effect destabilizes cellular homeostasis owing to the generation of Reactive Oxygen Species (ROS), resulting in the enhanced pro-inflammatory function of immunocytes. In our previous study, the Ce6-mediated photodynamic effect was found to have kept the viability of macrophages and to remodel them into the M1 phenotype. However, the mechanism remains unrevealed. The present study now explores the mechanism of photodynamic therapy (PDT)-mediated reprogramming of macrophages. As expected, Ce6-mediated PDT was capable of generating Reactive Oxygen Species, which was continuously degraded, causing "low intensity" damage to DNA and thereby triggering subsequent DNA damage response in macrophages. The Autophagy was thus observed in Ce6-treated macrophages and was shown to protect cells from being photodynamically apoptotic. More importantly, Ce6 PDT could activate the stimulator of interferon genes (STING) molecule, a sensor of DNA damage, which could activate the downstream nuclear factor kappa-B (NF-κB) upon activation, mediating the polarization of macrophages towards the M1 phenotype thereupon. In addition, inhibition of ROS induced by PDT attenuated the DNA damage, STING activation, and M1-phenotype reprogramming. Furthermore, the silence of the STING weakened Ce6 treatment-mediated M1 remodeling of macrophages as well. Altogether, these findings indicate the Ce6-induced photodynamic effect polarizes macrophages into an M1 phenotype through oxidative DNA damage and subsequent activation of the STING. This work reveals the crucial mechanism by which photodynamic therapy regulates the macrophage phenotype and also provides a novel intervenable signaling target for remodeling macrophages into the M1 phenotype.

Keywords

DNA damage response (DDR); STING molecule; autophagy; macrophages; photodynamic effect; reactive oxygen species (ROS).

Figures
Products