Cargando…

Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1

BACKGROUND: Cancer-associated fibroblasts (CAFs), the predominant stromal cell of tumor microenvironment (TME), play an important role in tumor progression and immunoregulation by remodeling extracellular matrix (ECM) and secreting cytokines. However, little is known about the details of the underly...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Zhao, Yu, Yongbo, Zhang, Zhilei, Li, Dan, Liang, Zhijuan, Wang, Liping, Chen, Yuanbin, Liang, Ye, Niu, Haitao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10675892/
https://www.ncbi.nlm.nih.gov/pubmed/38001512
http://dx.doi.org/10.1186/s13046-023-02900-0
_version_ 1785149869939228672
author Zhang, Zhao
Yu, Yongbo
Zhang, Zhilei
Li, Dan
Liang, Zhijuan
Wang, Liping
Chen, Yuanbin
Liang, Ye
Niu, Haitao
author_facet Zhang, Zhao
Yu, Yongbo
Zhang, Zhilei
Li, Dan
Liang, Zhijuan
Wang, Liping
Chen, Yuanbin
Liang, Ye
Niu, Haitao
author_sort Zhang, Zhao
collection PubMed
description BACKGROUND: Cancer-associated fibroblasts (CAFs), the predominant stromal cell of tumor microenvironment (TME), play an important role in tumor progression and immunoregulation by remodeling extracellular matrix (ECM) and secreting cytokines. However, little is known about the details of the underlying mechanism in bladder cancer. METHODS: Bioinformatics analysis was performed to analyze the prognostic value of CAFs and CXCL12 using GEO, TCGA and SRA databases. The effects of CXCL12 on bladder cancer progression were investigated through in vitro and in vivo assays. The biological mechanism of the effect of CXCL12 on PDL1 were investigated using western blotting, immunoprecipitation, RT-PCR, immunofluorescence, mass spectrometry, protein stability, and flow cytometry. RESULTS: The results demonstrated that CAFs-derived CXCL12 promoted cancer cell migration and invasion and upregulated PDL1. Mechanistically, upon binding to its specific receptor, CXCL12 activated the downstream JAK2/STAT3 pathway and rapidly up-regulated the expression of deubiquitinase CYLD. CYLD deubiquitinated P62 causing P62 accumulation, which in turn inhibited the autophagic degradation of PDL1. In vivo experiments demonstrated that blocking CXCL12 inhibited tumor growth, reduced tumor PDL1 expression and increased immune cell infiltration. CONCLUSIONS: This study revealed a novel mechanism for the role of CXCL12 in P62-mediated PDL1 autophagic regulation. Combined application of CXCL12 receptor blocker and PD1/PDL1 blocker can more effectively inhibit PDL1 expression and enhance antitumor immune response. Targeting CAFs-derived CXCL12 may provide an effective strategy for immunotherapy in bladder cancer. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02900-0.
format Online
Article
Text
id pubmed-10675892
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-106758922023-11-25 Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1 Zhang, Zhao Yu, Yongbo Zhang, Zhilei Li, Dan Liang, Zhijuan Wang, Liping Chen, Yuanbin Liang, Ye Niu, Haitao J Exp Clin Cancer Res Research BACKGROUND: Cancer-associated fibroblasts (CAFs), the predominant stromal cell of tumor microenvironment (TME), play an important role in tumor progression and immunoregulation by remodeling extracellular matrix (ECM) and secreting cytokines. However, little is known about the details of the underlying mechanism in bladder cancer. METHODS: Bioinformatics analysis was performed to analyze the prognostic value of CAFs and CXCL12 using GEO, TCGA and SRA databases. The effects of CXCL12 on bladder cancer progression were investigated through in vitro and in vivo assays. The biological mechanism of the effect of CXCL12 on PDL1 were investigated using western blotting, immunoprecipitation, RT-PCR, immunofluorescence, mass spectrometry, protein stability, and flow cytometry. RESULTS: The results demonstrated that CAFs-derived CXCL12 promoted cancer cell migration and invasion and upregulated PDL1. Mechanistically, upon binding to its specific receptor, CXCL12 activated the downstream JAK2/STAT3 pathway and rapidly up-regulated the expression of deubiquitinase CYLD. CYLD deubiquitinated P62 causing P62 accumulation, which in turn inhibited the autophagic degradation of PDL1. In vivo experiments demonstrated that blocking CXCL12 inhibited tumor growth, reduced tumor PDL1 expression and increased immune cell infiltration. CONCLUSIONS: This study revealed a novel mechanism for the role of CXCL12 in P62-mediated PDL1 autophagic regulation. Combined application of CXCL12 receptor blocker and PD1/PDL1 blocker can more effectively inhibit PDL1 expression and enhance antitumor immune response. Targeting CAFs-derived CXCL12 may provide an effective strategy for immunotherapy in bladder cancer. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02900-0. BioMed Central 2023-11-25 /pmc/articles/PMC10675892/ /pubmed/38001512 http://dx.doi.org/10.1186/s13046-023-02900-0 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, Zhao
Yu, Yongbo
Zhang, Zhilei
Li, Dan
Liang, Zhijuan
Wang, Liping
Chen, Yuanbin
Liang, Ye
Niu, Haitao
Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title_full Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title_fullStr Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title_full_unstemmed Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title_short Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1
title_sort cancer-associated fibroblasts-derived cxcl12 enhances immune escape of bladder cancer through inhibiting p62-mediated autophagic degradation of pdl1
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10675892/
https://www.ncbi.nlm.nih.gov/pubmed/38001512
http://dx.doi.org/10.1186/s13046-023-02900-0
work_keys_str_mv AT zhangzhao cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT yuyongbo cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT zhangzhilei cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT lidan cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT liangzhijuan cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT wangliping cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT chenyuanbin cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT liangye cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1
AT niuhaitao cancerassociatedfibroblastsderivedcxcl12enhancesimmuneescapeofbladdercancerthroughinhibitingp62mediatedautophagicdegradationofpdl1