Journal for ImmunoTherapy of Cancer (Mar 2023)

TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway

  • Yu Liu,
  • Zhe Zhang,
  • Min Luo,
  • Hao Zeng,
  • Yuquan Wei,
  • Siyuan Chen,
  • Xiaohe Tian,
  • Tianqi Lu,
  • Ziqi Zhang,
  • Zhenfei Bi,
  • Tianxia Lan,
  • Fei Mo,
  • Jingyun Yang,
  • Xuemei He,
  • Weiqi Hong,
  • Ruyu Pi,
  • Wenyan Ren,
  • Xiawei Wei

DOI
https://doi.org/10.1136/jitc-2022-005430
Journal volume & issue
Vol. 11, no. 3

Abstract

Read online

Background Mitochondrial transcription factor A (TFAM) is a transcription factor that maintains mitochondrial DNA (mtDNA) stabilization and initiates mtDNA replication. However, little is known about the immune regulation function and TFAM expression in immune cells in the tumors.Methods Mouse tumor models were applied to analyze the effect of TFAM deficiency in myeloid cell lineage on tumor progression and tumor microenvironment (TME) modification. In vitro, primary mouse bone marrow-derived dendritic cells (BMDCs) were used in the investigation of the altered function and the activated pathway. OVA was used as the model antigen to validate the activation of immune responses in vivo. STING inhibitors were used to confirm the STING activation provoked by Tfam deficient in DCs.Results The deletion of TFAM in DCs led to mitochondrial dysfunction and mtDNA cytosolic leakage resulting in the cGAS-STING pathway activation in DCs, which contributed to the enhanced antigen presentation. The deletion of TFAM in DCs has interestingly reversed the immune suppressive TME and inhibited tumor growth and metastasis in tumor models.Conclusions We have revealed that TFAM knockout in DCs ameliorated immune-suppressive microenvironment in tumors through STING pathway. Our work suggests that specific TFAM knockout in DCs might be a compelling strategy for designing novel immunotherapy methods in the future.