Cargando…

TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance

Immune checkpoint blockade (ICB) offers a new opportunity for treatment for gastric cancer (G.C.). Understanding the upstream regulation of immune checkpoints is crucial to further improve the efficacy of ICB therapy. Herein, using the CRISPR-Cas9-based genome-wide screening, we identified TRIM28 as...

Descripción completa

Detalles Bibliográficos
Autores principales: Ma, Xiaoxiao, Jia, Shuqin, Wang, Gangjian, Liang, Min, Guo, Ting, Du, Hong, Li, Sisi, Li, Xiaomei, Huangfu, Longtao, Guo, Jianping, Xing, Xiaofang, Ji, Jiafu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10290989/
https://www.ncbi.nlm.nih.gov/pubmed/37357254
http://dx.doi.org/10.1038/s41392-023-01450-3
_version_ 1785062603244961792
author Ma, Xiaoxiao
Jia, Shuqin
Wang, Gangjian
Liang, Min
Guo, Ting
Du, Hong
Li, Sisi
Li, Xiaomei
Huangfu, Longtao
Guo, Jianping
Xing, Xiaofang
Ji, Jiafu
author_facet Ma, Xiaoxiao
Jia, Shuqin
Wang, Gangjian
Liang, Min
Guo, Ting
Du, Hong
Li, Sisi
Li, Xiaomei
Huangfu, Longtao
Guo, Jianping
Xing, Xiaofang
Ji, Jiafu
author_sort Ma, Xiaoxiao
collection PubMed
description Immune checkpoint blockade (ICB) offers a new opportunity for treatment for gastric cancer (G.C.). Understanding the upstream regulation of immune checkpoints is crucial to further improve the efficacy of ICB therapy. Herein, using the CRISPR-Cas9-based genome-wide screening, we identified TRIM28 as one of the most significant regulators of PD-L1, a checkpoint protein, in G.C. cells. Mechanistically, TRIM28 directly binds to and stabilizes PD-L1 by inhibiting PD-L1 ubiquitination and promoting PD-L1 SUMOylation. Furthermore, TRIM28 facilitates K63 polyubiquitination of TBK1, activating TBK1-IRF1 and TBK1-mTOR pathways, resulting in enhanced PD-L1 transcription. It was found that TRIM28 was positively correlated with PD-L1 in G.C. cells. Moreover, high TRIM28 expression suggests poor survival in a cohort of 466 patients with G.C., and this observation is consistent while analyzing data from publicly available databases. Ectopic TRIM28 expression facilitated tumor growth, increased PD-L1 expression, and suppressed T cell activation in mice. Administration of the PD-L1 or TBK1 inhibitor significantly alleviated the TRIM28-induced tumor progression. Furthermore, combining the TBK1 inhibitor with CTLA4 immune checkpoint blockade has synergistic effects on G.C., and provides a novel strategy for G.C. therapy.
format Online
Article
Text
id pubmed-10290989
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-102909892023-06-27 TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance Ma, Xiaoxiao Jia, Shuqin Wang, Gangjian Liang, Min Guo, Ting Du, Hong Li, Sisi Li, Xiaomei Huangfu, Longtao Guo, Jianping Xing, Xiaofang Ji, Jiafu Signal Transduct Target Ther Article Immune checkpoint blockade (ICB) offers a new opportunity for treatment for gastric cancer (G.C.). Understanding the upstream regulation of immune checkpoints is crucial to further improve the efficacy of ICB therapy. Herein, using the CRISPR-Cas9-based genome-wide screening, we identified TRIM28 as one of the most significant regulators of PD-L1, a checkpoint protein, in G.C. cells. Mechanistically, TRIM28 directly binds to and stabilizes PD-L1 by inhibiting PD-L1 ubiquitination and promoting PD-L1 SUMOylation. Furthermore, TRIM28 facilitates K63 polyubiquitination of TBK1, activating TBK1-IRF1 and TBK1-mTOR pathways, resulting in enhanced PD-L1 transcription. It was found that TRIM28 was positively correlated with PD-L1 in G.C. cells. Moreover, high TRIM28 expression suggests poor survival in a cohort of 466 patients with G.C., and this observation is consistent while analyzing data from publicly available databases. Ectopic TRIM28 expression facilitated tumor growth, increased PD-L1 expression, and suppressed T cell activation in mice. Administration of the PD-L1 or TBK1 inhibitor significantly alleviated the TRIM28-induced tumor progression. Furthermore, combining the TBK1 inhibitor with CTLA4 immune checkpoint blockade has synergistic effects on G.C., and provides a novel strategy for G.C. therapy. Nature Publishing Group UK 2023-06-26 /pmc/articles/PMC10290989/ /pubmed/37357254 http://dx.doi.org/10.1038/s41392-023-01450-3 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Ma, Xiaoxiao
Jia, Shuqin
Wang, Gangjian
Liang, Min
Guo, Ting
Du, Hong
Li, Sisi
Li, Xiaomei
Huangfu, Longtao
Guo, Jianping
Xing, Xiaofang
Ji, Jiafu
TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title_full TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title_fullStr TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title_full_unstemmed TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title_short TRIM28 promotes the escape of gastric cancer cells from immune surveillance by increasing PD-L1 abundance
title_sort trim28 promotes the escape of gastric cancer cells from immune surveillance by increasing pd-l1 abundance
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10290989/
https://www.ncbi.nlm.nih.gov/pubmed/37357254
http://dx.doi.org/10.1038/s41392-023-01450-3
work_keys_str_mv AT maxiaoxiao trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT jiashuqin trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT wanggangjian trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT liangmin trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT guoting trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT duhong trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT lisisi trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT lixiaomei trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT huangfulongtao trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT guojianping trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT xingxiaofang trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance
AT jijiafu trim28promotestheescapeofgastriccancercellsfromimmunesurveillancebyincreasingpdl1abundance