Cargando…

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

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 tu...

Descripción completa

Detalles Bibliográficos
Autores principales: Lu, Tianqi, Zhang, Ziqi, Bi, Zhenfei, Lan, Tianxia, Zeng, Hao, Liu, Yu, Mo, Fei, Yang, Jingyun, Chen, Siyuan, He, Xuemei, Hong, Weiqi, Zhang, Zhe, Pi, Ruyu, Ren, Wenyan, Tian, Xiaohe, Wei, Yuquan, Luo, Min, Wei, Xiawei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9980377/
https://www.ncbi.nlm.nih.gov/pubmed/36858460
http://dx.doi.org/10.1136/jitc-2022-005430
_version_ 1784899903574507520
author Lu, Tianqi
Zhang, Ziqi
Bi, Zhenfei
Lan, Tianxia
Zeng, Hao
Liu, Yu
Mo, Fei
Yang, Jingyun
Chen, Siyuan
He, Xuemei
Hong, Weiqi
Zhang, Zhe
Pi, Ruyu
Ren, Wenyan
Tian, Xiaohe
Wei, Yuquan
Luo, Min
Wei, Xiawei
author_facet Lu, Tianqi
Zhang, Ziqi
Bi, Zhenfei
Lan, Tianxia
Zeng, Hao
Liu, Yu
Mo, Fei
Yang, Jingyun
Chen, Siyuan
He, Xuemei
Hong, Weiqi
Zhang, Zhe
Pi, Ruyu
Ren, Wenyan
Tian, Xiaohe
Wei, Yuquan
Luo, Min
Wei, Xiawei
author_sort Lu, Tianqi
collection PubMed
description 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.
format Online
Article
Text
id pubmed-9980377
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-99803772023-03-03 TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway Lu, Tianqi Zhang, Ziqi Bi, Zhenfei Lan, Tianxia Zeng, Hao Liu, Yu Mo, Fei Yang, Jingyun Chen, Siyuan He, Xuemei Hong, Weiqi Zhang, Zhe Pi, Ruyu Ren, Wenyan Tian, Xiaohe Wei, Yuquan Luo, Min Wei, Xiawei J Immunother Cancer Basic Tumor Immunology 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. BMJ Publishing Group 2023-03-01 /pmc/articles/PMC9980377/ /pubmed/36858460 http://dx.doi.org/10.1136/jitc-2022-005430 Text en © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Basic Tumor Immunology
Lu, Tianqi
Zhang, Ziqi
Bi, Zhenfei
Lan, Tianxia
Zeng, Hao
Liu, Yu
Mo, Fei
Yang, Jingyun
Chen, Siyuan
He, Xuemei
Hong, Weiqi
Zhang, Zhe
Pi, Ruyu
Ren, Wenyan
Tian, Xiaohe
Wei, Yuquan
Luo, Min
Wei, Xiawei
TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title_full TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title_fullStr TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title_full_unstemmed TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title_short TFAM deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cGAS-STING pathway
title_sort tfam deficiency in dendritic cells leads to mitochondrial dysfunction and enhanced antitumor immunity through cgas-sting pathway
topic Basic Tumor Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9980377/
https://www.ncbi.nlm.nih.gov/pubmed/36858460
http://dx.doi.org/10.1136/jitc-2022-005430
work_keys_str_mv AT lutianqi tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT zhangziqi tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT bizhenfei tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT lantianxia tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT zenghao tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT liuyu tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT mofei tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT yangjingyun tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT chensiyuan tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT hexuemei tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT hongweiqi tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT zhangzhe tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT piruyu tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT renwenyan tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT tianxiaohe tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT weiyuquan tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT luomin tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway
AT weixiawei tfamdeficiencyindendriticcellsleadstomitochondrialdysfunctionandenhancedantitumorimmunitythroughcgasstingpathway