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

C Li, H Chi, S Deng, K Xu, H Wang, H Yao… - … for immunotherapy of …, 2021 - ncbi.nlm.nih.gov
C Li, H Chi, S Deng, K Xu, H Wang, H Yao, Y Wang, D Chen, X Guo, JY Fang, F He, J Xu
Journal for immunotherapy of cancer, 2021ncbi.nlm.nih.gov
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 …
Abstract
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.
ncbi.nlm.nih.gov