Cargando…

PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination

Programmed death-ligand 1 (PD-L1), a critical immune checkpoint ligand, is a transmembrane protein synthesized in the endoplasmic reticulum of tumor cells and transported to the plasma membrane to interact with programmed death 1 (PD-1) expressed on T cell surface. This interaction delivers coinhibi...

Descripción completa

Detalles Bibliográficos
Autores principales: Yu, Xinfang, Li, Wei, Liu, Haidan, Wang, Xu, Coarfa, Cristian, Cheng, Chao, Yu, Xinlian, Zeng, Zhaoyang, Cao, Ya, Young, Ken H., Li, Yong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Clinical Investigation 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9888393/
https://www.ncbi.nlm.nih.gov/pubmed/36719382
http://dx.doi.org/10.1172/JCI160456
_version_ 1784880524291997696
author Yu, Xinfang
Li, Wei
Liu, Haidan
Wang, Xu
Coarfa, Cristian
Cheng, Chao
Yu, Xinlian
Zeng, Zhaoyang
Cao, Ya
Young, Ken H.
Li, Yong
author_facet Yu, Xinfang
Li, Wei
Liu, Haidan
Wang, Xu
Coarfa, Cristian
Cheng, Chao
Yu, Xinlian
Zeng, Zhaoyang
Cao, Ya
Young, Ken H.
Li, Yong
author_sort Yu, Xinfang
collection PubMed
description Programmed death-ligand 1 (PD-L1), a critical immune checkpoint ligand, is a transmembrane protein synthesized in the endoplasmic reticulum of tumor cells and transported to the plasma membrane to interact with programmed death 1 (PD-1) expressed on T cell surface. This interaction delivers coinhibitory signals to T cells, thereby suppressing their function and allowing evasion of antitumor immunity. Most companion or complementary diagnostic devices for assessing PD-L1 expression levels in tumor cells used in the clinic or in clinical trials require membranous staining. However, the mechanism driving PD-L1 translocation to the plasma membrane after de novo synthesis is poorly understood. Herein, we showed that mind bomb homolog 2 (MIB2) is required for PD-L1 transportation from the trans-Golgi network (TGN) to the plasma membrane of cancer cells. MIB2 deficiency led to fewer PD-L1 proteins on the tumor cell surface and promoted antitumor immunity in mice. Mechanistically, MIB2 catalyzed nonproteolytic K63-linked ubiquitination of PD-L1, facilitating PD-L1 trafficking through Ras-associated binding 8–mediated (RAB8-mediated) exocytosis from the TGN to the plasma membrane, where it bound PD-1 extrinsically to prevent tumor cell killing by T cells. Our findings demonstrate that nonproteolytic ubiquitination of PD-L1 by MIB2 is required for its transportation to the plasma membrane and tumor cell immune evasion.
format Online
Article
Text
id pubmed-9888393
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher American Society for Clinical Investigation
record_format MEDLINE/PubMed
spelling pubmed-98883932023-02-06 PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination Yu, Xinfang Li, Wei Liu, Haidan Wang, Xu Coarfa, Cristian Cheng, Chao Yu, Xinlian Zeng, Zhaoyang Cao, Ya Young, Ken H. Li, Yong J Clin Invest Research Article Programmed death-ligand 1 (PD-L1), a critical immune checkpoint ligand, is a transmembrane protein synthesized in the endoplasmic reticulum of tumor cells and transported to the plasma membrane to interact with programmed death 1 (PD-1) expressed on T cell surface. This interaction delivers coinhibitory signals to T cells, thereby suppressing their function and allowing evasion of antitumor immunity. Most companion or complementary diagnostic devices for assessing PD-L1 expression levels in tumor cells used in the clinic or in clinical trials require membranous staining. However, the mechanism driving PD-L1 translocation to the plasma membrane after de novo synthesis is poorly understood. Herein, we showed that mind bomb homolog 2 (MIB2) is required for PD-L1 transportation from the trans-Golgi network (TGN) to the plasma membrane of cancer cells. MIB2 deficiency led to fewer PD-L1 proteins on the tumor cell surface and promoted antitumor immunity in mice. Mechanistically, MIB2 catalyzed nonproteolytic K63-linked ubiquitination of PD-L1, facilitating PD-L1 trafficking through Ras-associated binding 8–mediated (RAB8-mediated) exocytosis from the TGN to the plasma membrane, where it bound PD-1 extrinsically to prevent tumor cell killing by T cells. Our findings demonstrate that nonproteolytic ubiquitination of PD-L1 by MIB2 is required for its transportation to the plasma membrane and tumor cell immune evasion. American Society for Clinical Investigation 2023-02-01 /pmc/articles/PMC9888393/ /pubmed/36719382 http://dx.doi.org/10.1172/JCI160456 Text en © 2023 Yu et al. https://creativecommons.org/licenses/by/4.0/This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Yu, Xinfang
Li, Wei
Liu, Haidan
Wang, Xu
Coarfa, Cristian
Cheng, Chao
Yu, Xinlian
Zeng, Zhaoyang
Cao, Ya
Young, Ken H.
Li, Yong
PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title_full PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title_fullStr PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title_full_unstemmed PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title_short PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination
title_sort pd-l1 translocation to the plasma membrane enables tumor immune evasion through mib2 ubiquitination
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9888393/
https://www.ncbi.nlm.nih.gov/pubmed/36719382
http://dx.doi.org/10.1172/JCI160456
work_keys_str_mv AT yuxinfang pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT liwei pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT liuhaidan pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT wangxu pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT coarfacristian pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT chengchao pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT yuxinlian pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT zengzhaoyang pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT caoya pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT youngkenh pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination
AT liyong pdl1translocationtotheplasmamembraneenablestumorimmuneevasionthroughmib2ubiquitination