Cargando…

UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer

Background: The up-regulation of PD-L1 is recognized as an adaption of cancer cells to evade immune surveillance and attack. However, the intrinsic mechanisms of the induction of PD-L1 by interferon-γ (IFN-γ) in tumor microenvironment remain incompletely characterized. Ubiquitin ligase E3 component...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Bingbing, Song, Mei, Dong, Qun, Xiang, Gang, Li, Jing, Ma, Xiaojing, Wei, Fang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9274738/
https://www.ncbi.nlm.nih.gov/pubmed/35836797
http://dx.doi.org/10.7150/thno.74989
_version_ 1784745352353546240
author Wu, Bingbing
Song, Mei
Dong, Qun
Xiang, Gang
Li, Jing
Ma, Xiaojing
Wei, Fang
author_facet Wu, Bingbing
Song, Mei
Dong, Qun
Xiang, Gang
Li, Jing
Ma, Xiaojing
Wei, Fang
author_sort Wu, Bingbing
collection PubMed
description Background: The up-regulation of PD-L1 is recognized as an adaption of cancer cells to evade immune surveillance and attack. However, the intrinsic mechanisms of the induction of PD-L1 by interferon-γ (IFN-γ) in tumor microenvironment remain incompletely characterized. Ubiquitin ligase E3 component N-recognition protein 5 (UBR5) has a critical role in tumorigenesis of triple negative breast cancer (TNBC) by triggering specific immune responses to the tumor. Dual targeting of UBR5 and PD-L1 exhibited superior therapeutic benefits in a preclinical TNBC model in short term. Methods: The regulation of UBR5 to PD-L1 upon IFN-γ stimulation was evaluated through in UBR5 deficiency, reconstitution or overexpression cell line models by quantitative PCR, immunohistochemistry and RNA-seq. The effects of PD-L1 regulation by UBR5 and double blockade of both genes were evaluated in mouse TNBC model. Luciferase reporter assay, chromatin immunoprecipitation-qPCR and bioinformatics analysis were performed to explore the transcription factors involved in the regulation of UBR5 to PD-L1. Results: E3 ubiquitin ligase UBR5 plays a key role in IFN-γ-induced PDL1 transcription in TNBC in an E3 ubiquitination activity-independent manner. RNA-seq-based transcriptomic analyses reveal that UBR5 globally affects the genes in the IFN-γ-induced signaling pathway. Through its poly adenylate binding (PABC) domain, UBR5 enhances the transactivation of PDL1 by upregulating protein kinase RNA-activated (PKR), and PKR's downstream factors including signal transducers and activators of transcription 1 (STAT1) and interferon regulatory factor 1 (IRF1). Restoration of PD-L1 expression in UBR5-deficient tumor cells recoups their malignancy in vivo, whereas CRISPR/Cas9-mediated simultaneous abrogation of UBR5 and PD-L1 expression yields synergistic therapeutic benefits than either blockade alone, with a strong impact on the tumor microenvironment. Conclusions: This study identifies a novel regulator of PDL1 transcription, elucidates the underlying molecular mechanisms and provides a strong rationale for combination cancer immunotherapies targeting UBR5 and PD-L1.
format Online
Article
Text
id pubmed-9274738
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-92747382022-07-13 UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer Wu, Bingbing Song, Mei Dong, Qun Xiang, Gang Li, Jing Ma, Xiaojing Wei, Fang Theranostics Research Paper Background: The up-regulation of PD-L1 is recognized as an adaption of cancer cells to evade immune surveillance and attack. However, the intrinsic mechanisms of the induction of PD-L1 by interferon-γ (IFN-γ) in tumor microenvironment remain incompletely characterized. Ubiquitin ligase E3 component N-recognition protein 5 (UBR5) has a critical role in tumorigenesis of triple negative breast cancer (TNBC) by triggering specific immune responses to the tumor. Dual targeting of UBR5 and PD-L1 exhibited superior therapeutic benefits in a preclinical TNBC model in short term. Methods: The regulation of UBR5 to PD-L1 upon IFN-γ stimulation was evaluated through in UBR5 deficiency, reconstitution or overexpression cell line models by quantitative PCR, immunohistochemistry and RNA-seq. The effects of PD-L1 regulation by UBR5 and double blockade of both genes were evaluated in mouse TNBC model. Luciferase reporter assay, chromatin immunoprecipitation-qPCR and bioinformatics analysis were performed to explore the transcription factors involved in the regulation of UBR5 to PD-L1. Results: E3 ubiquitin ligase UBR5 plays a key role in IFN-γ-induced PDL1 transcription in TNBC in an E3 ubiquitination activity-independent manner. RNA-seq-based transcriptomic analyses reveal that UBR5 globally affects the genes in the IFN-γ-induced signaling pathway. Through its poly adenylate binding (PABC) domain, UBR5 enhances the transactivation of PDL1 by upregulating protein kinase RNA-activated (PKR), and PKR's downstream factors including signal transducers and activators of transcription 1 (STAT1) and interferon regulatory factor 1 (IRF1). Restoration of PD-L1 expression in UBR5-deficient tumor cells recoups their malignancy in vivo, whereas CRISPR/Cas9-mediated simultaneous abrogation of UBR5 and PD-L1 expression yields synergistic therapeutic benefits than either blockade alone, with a strong impact on the tumor microenvironment. Conclusions: This study identifies a novel regulator of PDL1 transcription, elucidates the underlying molecular mechanisms and provides a strong rationale for combination cancer immunotherapies targeting UBR5 and PD-L1. Ivyspring International Publisher 2022-07-04 /pmc/articles/PMC9274738/ /pubmed/35836797 http://dx.doi.org/10.7150/thno.74989 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Wu, Bingbing
Song, Mei
Dong, Qun
Xiang, Gang
Li, Jing
Ma, Xiaojing
Wei, Fang
UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title_full UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title_fullStr UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title_full_unstemmed UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title_short UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer
title_sort ubr5 promotes tumor immune evasion through enhancing ifn-γ-induced pdl1 transcription in triple negative breast cancer
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9274738/
https://www.ncbi.nlm.nih.gov/pubmed/35836797
http://dx.doi.org/10.7150/thno.74989
work_keys_str_mv AT wubingbing ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT songmei ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT dongqun ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT xianggang ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT lijing ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT maxiaojing ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer
AT weifang ubr5promotestumorimmuneevasionthroughenhancingifnginducedpdl1transcriptionintriplenegativebreastcancer