Cargando…

THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy

BACKGROUND: The abnormal upregulation of programmed death-ligand 1 (PD-L1) in cancer cells inhibits T cell-mediated cytotoxicity, but the molecular mechanisms that drive and maintain PD-L1 expression are still incompletely understood. METHODS: Combined analyses of genomes and proteomics were applied...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Chushu, Chi, Hao, Deng, Shouyan, Xu, Ke, Wang, Huanbin, Yao, Han, Wang, Yungang, Chen, Dawei, Guo, Xun, Fang, Jing-Yuan, He, Fang, Xu, Jie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8330570/
https://www.ncbi.nlm.nih.gov/pubmed/34341130
http://dx.doi.org/10.1136/jitc-2021-002443
_version_ 1783732746695213056
author Li, Chushu
Chi, Hao
Deng, Shouyan
Xu, Ke
Wang, Huanbin
Yao, Han
Wang, Yungang
Chen, Dawei
Guo, Xun
Fang, Jing-Yuan
He, Fang
Xu, Jie
author_facet Li, Chushu
Chi, Hao
Deng, Shouyan
Xu, Ke
Wang, Huanbin
Yao, Han
Wang, Yungang
Chen, Dawei
Guo, Xun
Fang, Jing-Yuan
He, Fang
Xu, Jie
author_sort Li, Chushu
collection PubMed
description BACKGROUND: The abnormal upregulation of programmed death-ligand 1 (PD-L1) in cancer cells inhibits T cell-mediated cytotoxicity, but the molecular mechanisms that drive and maintain PD-L1 expression are still incompletely understood. METHODS: Combined analyses of genomes and proteomics were applied to find potential regulators of PD-L1. In vitro experiments were performed to investigate the regulatory mechanism of PD-L1 by thyroid adenoma associated gene (THADA) using human colorectal cancer (CRC) cells. The prevalence of THADA was analyzed using CRC tissue microarrays by immunohistochemistry. T cell killing assay, programmed cell death 1 binding assay and MC38 transplanted tumor models in C57BL/6 mice were developed to investigate the antitumor effect of THADA. RESULTS: THADA is critically required for the Golgi residency of PD-L1, and this non-redundant, coat protein complex II (COPII)-associated mechanism maintains PD-L1 expression in tumor cells. THADA mediated the interaction between PD-L1 as a cargo protein with SEC24A, a module on the COPII trafficking vesicle. Silencing THADA caused absence and endoplasmic reticulum (ER) retention of PD-L1 but not major histocompatibility complex-I, inducing PD-L1 clearance through ER-associated degradation. Targeting THADA substantially enhanced T cell-mediated cytotoxicity, and increased CD8+ T cells infiltration in mouse tumor tissues. Analysis on clinical tissue samples supported a potential role of THADA in upregulating PD-L1 expression in cancer. CONCLUSIONS: Our data reveal a crucial cellular process for PD-L1 maturation and maintenance in tumor cells, and highlight THADA as a promising target for overcoming PD-L1-dependent immune evasion.
format Online
Article
Text
id pubmed-8330570
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-83305702021-08-20 THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy Li, Chushu Chi, Hao Deng, Shouyan Xu, Ke Wang, Huanbin Yao, Han Wang, Yungang Chen, Dawei Guo, Xun Fang, Jing-Yuan He, Fang Xu, Jie J Immunother Cancer Basic Tumor Immunology BACKGROUND: The abnormal upregulation of programmed death-ligand 1 (PD-L1) in cancer cells inhibits T cell-mediated cytotoxicity, but the molecular mechanisms that drive and maintain PD-L1 expression are still incompletely understood. METHODS: Combined analyses of genomes and proteomics were applied to find potential regulators of PD-L1. In vitro experiments were performed to investigate the regulatory mechanism of PD-L1 by thyroid adenoma associated gene (THADA) using human colorectal cancer (CRC) cells. The prevalence of THADA was analyzed using CRC tissue microarrays by immunohistochemistry. T cell killing assay, programmed cell death 1 binding assay and MC38 transplanted tumor models in C57BL/6 mice were developed to investigate the antitumor effect of THADA. RESULTS: THADA is critically required for the Golgi residency of PD-L1, and this non-redundant, coat protein complex II (COPII)-associated mechanism maintains PD-L1 expression in tumor cells. THADA mediated the interaction between PD-L1 as a cargo protein with SEC24A, a module on the COPII trafficking vesicle. Silencing THADA caused absence and endoplasmic reticulum (ER) retention of PD-L1 but not major histocompatibility complex-I, inducing PD-L1 clearance through ER-associated degradation. Targeting THADA substantially enhanced T cell-mediated cytotoxicity, and increased CD8+ T cells infiltration in mouse tumor tissues. Analysis on clinical tissue samples supported a potential role of THADA in upregulating PD-L1 expression in cancer. CONCLUSIONS: Our data reveal a crucial cellular process for PD-L1 maturation and maintenance in tumor cells, and highlight THADA as a promising target for overcoming PD-L1-dependent immune evasion. BMJ Publishing Group 2021-08-02 /pmc/articles/PMC8330570/ /pubmed/34341130 http://dx.doi.org/10.1136/jitc-2021-002443 Text en © Author(s) (or their employer(s)) 2021. 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
Li, Chushu
Chi, Hao
Deng, Shouyan
Xu, Ke
Wang, Huanbin
Yao, Han
Wang, Yungang
Chen, Dawei
Guo, Xun
Fang, Jing-Yuan
He, Fang
Xu, Jie
THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title_full THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title_fullStr THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title_full_unstemmed THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title_short THADA drives Golgi residency and upregulation of PD-L1 in cancer cells and provides promising target for immunotherapy
title_sort thada drives golgi residency and upregulation of pd-l1 in cancer cells and provides promising target for immunotherapy
topic Basic Tumor Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8330570/
https://www.ncbi.nlm.nih.gov/pubmed/34341130
http://dx.doi.org/10.1136/jitc-2021-002443
work_keys_str_mv AT lichushu thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT chihao thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT dengshouyan thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT xuke thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT wanghuanbin thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT yaohan thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT wangyungang thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT chendawei thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT guoxun thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT fangjingyuan thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT hefang thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy
AT xujie thadadrivesgolgiresidencyandupregulationofpdl1incancercellsandprovidespromisingtargetforimmunotherapy