Cargando…

Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling

BACKGROUND: Immune checkpoint blockade (ICB) represents a revolutionary advance in cancer treatment but remains limited success in triple-negative breast cancer (TNBC). Here we aim to explore the mechanism of RNA-binding protein (RBP) HuR in cancer immune evasion by post-transcriptionally regulating...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Qi, Yang, Zhe, Hao, Xinbao, Dandreo, Lauren J., He, Lily, Zhang, Yuxia, Wang, Fen, Wu, Xiaoqing, Xu, Liang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10583380/
https://www.ncbi.nlm.nih.gov/pubmed/37848943
http://dx.doi.org/10.1186/s13578-023-01137-w
_version_ 1785122538963075072
author Zhang, Qi
Yang, Zhe
Hao, Xinbao
Dandreo, Lauren J.
He, Lily
Zhang, Yuxia
Wang, Fen
Wu, Xiaoqing
Xu, Liang
author_facet Zhang, Qi
Yang, Zhe
Hao, Xinbao
Dandreo, Lauren J.
He, Lily
Zhang, Yuxia
Wang, Fen
Wu, Xiaoqing
Xu, Liang
author_sort Zhang, Qi
collection PubMed
description BACKGROUND: Immune checkpoint blockade (ICB) represents a revolutionary advance in cancer treatment but remains limited success in triple-negative breast cancer (TNBC). Here we aim to explore the mechanism of RNA-binding protein (RBP) HuR in cancer immune evasion by post-transcriptionally regulating PD-L1 and evaluate the potential of HuR inhibition to improve immune response. METHODS: The binding between HuR and PD-L1 mRNA was determined by ribonucleoprotein immunoprecipitation and RNA pull-down assays. The HuR knockout clones were established by CRISPR/Cas9 technology. The protein levels were assessed by Western blot, immunohistochemistry, and immunocytochemistry. The function and molecular mechanism of HuR-PD-L1 were determined by in vitro T cell activation and killing assay and in vivo efficacy assay. RESULTS: We found that HuR directly bound to and stabilized PD-L1 mRNA. Knocking out HuR reduced PD-L1 levels and promoted T cell activation. We discovered that niclosamide reduced PD-L1 by inhibiting HuR cytoplasmic translocation, and diminished glycosylation of PD-L1. Niclosamide enhanced T cell-mediated killing of cancer cells and significantly improved the efficacy of anti-PD-1 immunotherapy in two syngeneic animal tumor models. CONCLUSION: We identified HuR as a novel posttranscriptional regulator of PD-L1, which plays an important role in tumor immune evasion. Niclosamide might be a promising repurposed drug to improve the patient response to immunotherapy by targeting HuR-PD-L1 axis. Our study demonstrates a novel strategy for targeting HuR/PD-L1 and provides the first proof-of-principle for repurposing niclosamide as a HuR inhibitor to overcome cancer immune evasion and improve response to ICB immunotherapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01137-w.
format Online
Article
Text
id pubmed-10583380
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105833802023-10-19 Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling Zhang, Qi Yang, Zhe Hao, Xinbao Dandreo, Lauren J. He, Lily Zhang, Yuxia Wang, Fen Wu, Xiaoqing Xu, Liang Cell Biosci Research BACKGROUND: Immune checkpoint blockade (ICB) represents a revolutionary advance in cancer treatment but remains limited success in triple-negative breast cancer (TNBC). Here we aim to explore the mechanism of RNA-binding protein (RBP) HuR in cancer immune evasion by post-transcriptionally regulating PD-L1 and evaluate the potential of HuR inhibition to improve immune response. METHODS: The binding between HuR and PD-L1 mRNA was determined by ribonucleoprotein immunoprecipitation and RNA pull-down assays. The HuR knockout clones were established by CRISPR/Cas9 technology. The protein levels were assessed by Western blot, immunohistochemistry, and immunocytochemistry. The function and molecular mechanism of HuR-PD-L1 were determined by in vitro T cell activation and killing assay and in vivo efficacy assay. RESULTS: We found that HuR directly bound to and stabilized PD-L1 mRNA. Knocking out HuR reduced PD-L1 levels and promoted T cell activation. We discovered that niclosamide reduced PD-L1 by inhibiting HuR cytoplasmic translocation, and diminished glycosylation of PD-L1. Niclosamide enhanced T cell-mediated killing of cancer cells and significantly improved the efficacy of anti-PD-1 immunotherapy in two syngeneic animal tumor models. CONCLUSION: We identified HuR as a novel posttranscriptional regulator of PD-L1, which plays an important role in tumor immune evasion. Niclosamide might be a promising repurposed drug to improve the patient response to immunotherapy by targeting HuR-PD-L1 axis. Our study demonstrates a novel strategy for targeting HuR/PD-L1 and provides the first proof-of-principle for repurposing niclosamide as a HuR inhibitor to overcome cancer immune evasion and improve response to ICB immunotherapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01137-w. BioMed Central 2023-10-17 /pmc/articles/PMC10583380/ /pubmed/37848943 http://dx.doi.org/10.1186/s13578-023-01137-w 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Zhang, Qi
Yang, Zhe
Hao, Xinbao
Dandreo, Lauren J.
He, Lily
Zhang, Yuxia
Wang, Fen
Wu, Xiaoqing
Xu, Liang
Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title_full Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title_fullStr Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title_full_unstemmed Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title_short Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling
title_sort niclosamide improves cancer immunotherapy by modulating rna-binding protein hur-mediated pd-l1 signaling
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10583380/
https://www.ncbi.nlm.nih.gov/pubmed/37848943
http://dx.doi.org/10.1186/s13578-023-01137-w
work_keys_str_mv AT zhangqi niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT yangzhe niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT haoxinbao niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT dandreolaurenj niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT helily niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT zhangyuxia niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT wangfen niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT wuxiaoqing niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling
AT xuliang niclosamideimprovescancerimmunotherapybymodulatingrnabindingproteinhurmediatedpdl1signaling