Skip to main content
Top

30-04-2024 | Esophageal Cancer | RESEARCH

FOXP4-AS1 promotes CD8+ T cell exhaustion and esophageal cancer immune escape through USP10-stabilized PD-L1

Authors: Guo-yi Shen, Yi Zhang, Rong-zhi Huang, Zhi-yong Huang, Le-yi Yang, Ding-zhu Chen, Shao-bin Yang

Published in: Immunologic Research

Login to get access

Abstract

Esophageal cancer (EC) is the 9th most frequently diagnosed malignancy globally with unfavorable prognosis. Immune escape is one of the principal factors leading to poor survival, however, the mechanism underlying immune escape remains largely uninvestigated. The xenograft mouse model and EC cell-CD8+ cytotoxic T lymphocytes (CTLs) co-culture system were established. Immunohistochemistry, qRT-PCR or western blot were employed to detect the levels of long non-coding RNA (lncRNA) FOXP4-AS1, PD-L1, USP10 and other molecules. The abundance of T cells, cytokine production and cell apoptosis were monitored by flow cytometry. The viability of CTLs was assessed by Trypan blue staining. The binding between FOXP4-AS1 and USP10 was validated by RNA pull-down assay, and the interaction between USP10 and PD-L1, as well as the ubiquitination of PD-L1, were detected by co-immunoprecipitation. The elevation of FOXP4-AS1 in EC was associated with decreased CTL abundance, and upregulated PD-L1 facilitated CTL apoptosis in EC. FOXP4-AS1 accelerated EC tumor growth by decreasing the abundance of tumor infiltrating CTLs in vivo. FOXP4-AS1 inhibited the viability of CTLs and facilitated the cytotoxicity and exhaustion of CTLs. In Kyse 450 cell-CTL co-culture system, FOXP4-AS1 suppressed the viability and abundance of CTLs, and inhibited EC cell apoptosis via PD-L1. Mechanistically, FOXP4-AS1 regulated the ubiquitination of PD-L1 through deubiquitinating enzyme USP10. FOXP4-AS1 promoted CTL exhaustion and EC immune escape through USP10-stabilized PD-L1.

Highlights

  • PD-L1 facilitated CD8+ T cell apoptosis in EC.
  • Upregulated FOXP4-AS1 promoted EC tumor growth by inhibiting the viability and facilitating the cytotoxicity and exhaustion of tumor infiltrating CD8+ T cells.
  • FOXP4-AS1 suppressed the viability and abundance of CD8+ T cells through USP10-mediated deubiquitination of PD-L1.
Literature
1.
go back to reference Sheikh M, et al. Current status and future prospects for esophageal cancer. Cancers. 2023;15(3):765. Sheikh M, et al. Current status and future prospects for esophageal cancer. Cancers. 2023;15(3):765.
2.
3.
5.
7.
go back to reference Li R, et al. Immune evasion in esophageal squamous cell cancer: From the perspective of tumor microenvironment. Front Oncol. 2022;12:1096717.CrossRefPubMed Li R, et al. Immune evasion in esophageal squamous cell cancer: From the perspective of tumor microenvironment. Front Oncol. 2022;12:1096717.CrossRefPubMed
8.
go back to reference Vrana D, et al. From tumor immunology to immunotherapy in gastric and esophageal cancer. Int J Mol Sci. 2018;20(1):13. Vrana D, et al. From tumor immunology to immunotherapy in gastric and esophageal cancer. Int J Mol Sci. 2018;20(1):13.
10.
go back to reference Zhang G, et al. FOXP4-AS1 may be a potential prognostic biomarker in human cancers: a meta-analysis and bioinformatics analysis. Front Oncol. 2022;12:799265.CrossRefPubMedPubMedCentral Zhang G, et al. FOXP4-AS1 may be a potential prognostic biomarker in human cancers: a meta-analysis and bioinformatics analysis. Front Oncol. 2022;12:799265.CrossRefPubMedPubMedCentral
11.
go back to reference Niu Y, et al. LncRNA FOXP4-AS1 promotes the progression of esophageal squamous cell carcinoma by interacting with MLL2/H3K4me3 to upregulate FOXP4. Front Oncol. 2021;11:773864.CrossRefPubMedPubMedCentral Niu Y, et al. LncRNA FOXP4-AS1 promotes the progression of esophageal squamous cell carcinoma by interacting with MLL2/H3K4me3 to upregulate FOXP4. Front Oncol. 2021;11:773864.CrossRefPubMedPubMedCentral
12.
go back to reference Li Y, et al. YY1-induced upregulation of FOXP4-AS1 and FOXP4 promote the proliferation of esophageal squamous cell carcinoma cells. Cell Biol Int. 2020;44(7):1447–57.CrossRefPubMed Li Y, et al. YY1-induced upregulation of FOXP4-AS1 and FOXP4 promote the proliferation of esophageal squamous cell carcinoma cells. Cell Biol Int. 2020;44(7):1447–57.CrossRefPubMed
13.
go back to reference Jiang X, Chen ZJ. The role of ubiquitylation in immune defence and pathogen evasion. Nat Rev Immunol. 2011;12(1):35–48.CrossRefPubMed Jiang X, Chen ZJ. The role of ubiquitylation in immune defence and pathogen evasion. Nat Rev Immunol. 2011;12(1):35–48.CrossRefPubMed
16.
go back to reference Cao YF, et al. Targeting USP10 induces degradation of oncogenic ANLN in esophageal squamous cell carcinoma. Cell Death Differ. 2023;30(2):527–43.CrossRefPubMed Cao YF, et al. Targeting USP10 induces degradation of oncogenic ANLN in esophageal squamous cell carcinoma. Cell Death Differ. 2023;30(2):527–43.CrossRefPubMed
17.
go back to reference Borgmann M, Quante M. Impact of the tumor microenvironment for esophageal tumor development-an opportunity for prevention?. Cancers. 2022;14(9):2246. Borgmann M, Quante M. Impact of the tumor microenvironment for esophageal tumor development-an opportunity for prevention?. Cancers. 2022;14(9):2246.
18.
19.
go back to reference Liao C, et al. Long non-coding RNA FOXP4-AS1 is a prognostic biomarker and associated with immune infiltrates in ovarian serous cystadenocarcinoma. Medicine. 2021;100(40):e27473.CrossRefPubMedPubMedCentral Liao C, et al. Long non-coding RNA FOXP4-AS1 is a prognostic biomarker and associated with immune infiltrates in ovarian serous cystadenocarcinoma. Medicine. 2021;100(40):e27473.CrossRefPubMedPubMedCentral
21.
go back to reference Tao L, et al. USP10 as a potential therapeutic target in human cancers. Genes. 2022;13(5):831. Tao L, et al. USP10 as a potential therapeutic target in human cancers. Genes. 2022;13(5):831.
22.
24.
go back to reference Chen L, et al. PD-L1 expression promotes epithelial to mesenchymal transition in human esophageal cancer. Cell Physiol Biochem. 2017;42(6):2267–80.CrossRefPubMed Chen L, et al. PD-L1 expression promotes epithelial to mesenchymal transition in human esophageal cancer. Cell Physiol Biochem. 2017;42(6):2267–80.CrossRefPubMed
25.
go back to reference Xu J, et al. Evidence that dysplasia related microRNAs in Barrett’s esophagus target PD-L1 expression and contribute to the development of esophageal adenocarcinoma. Aging. 2020;12(17):17062–78.CrossRefPubMedPubMedCentral Xu J, et al. Evidence that dysplasia related microRNAs in Barrett’s esophagus target PD-L1 expression and contribute to the development of esophageal adenocarcinoma. Aging. 2020;12(17):17062–78.CrossRefPubMedPubMedCentral
26.
go back to reference Wang H, et al. Exosomal PD-L1 confers chemoresistance and promotes tumorigenic properties in esophageal cancer cells via upregulating STAT3/miR-21. Gene Ther. 2023;30(1–2):88–100.CrossRefPubMed Wang H, et al. Exosomal PD-L1 confers chemoresistance and promotes tumorigenic properties in esophageal cancer cells via upregulating STAT3/miR-21. Gene Ther. 2023;30(1–2):88–100.CrossRefPubMed
Metadata
Title
FOXP4-AS1 promotes CD8+ T cell exhaustion and esophageal cancer immune escape through USP10-stabilized PD-L1
Authors
Guo-yi Shen
Yi Zhang
Rong-zhi Huang
Zhi-yong Huang
Le-yi Yang
Ding-zhu Chen
Shao-bin Yang
Publication date
30-04-2024
Publisher
Springer US
Published in
Immunologic Research
Print ISSN: 0257-277X
Electronic ISSN: 1559-0755
DOI
https://doi.org/10.1007/s12026-024-09482-9