Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Mar 4:12:815437.
doi: 10.3389/fonc.2022.815437. eCollection 2022.

TGF-β1-Mediated PD-L1 Glycosylation Contributes to Immune Escape via c-Jun/STT3A Pathway in Nasopharyngeal Carcinoma

Affiliations

TGF-β1-Mediated PD-L1 Glycosylation Contributes to Immune Escape via c-Jun/STT3A Pathway in Nasopharyngeal Carcinoma

Xue-Min Ma et al. Front Oncol. .

Abstract

Immunotherapy targeting programmed death ligand-1/programmed cell death protein-1 (PD-L1/PD-1) has achieved great success in multiple cancers, but only a small subset of patients showed clinical responses. Recent evidences have shown that post-translational modification of PD-L1 protein could regulate its protein stability and interaction with cognate receptor PD-1, thereby affecting anticancer immunotherapy in several solid tumors. However, the molecular mechanisms underlying how PD-1/PD-L1 expression is regulated still remain unclear in nasopharyngeal carcinoma (NPC). Here, we found N-glycosylation of PD-L1 in NPC cells and tissues. Mechanistically, we showed that STT3A transferred N-linked glycans to PD-L1, and TGF-β1 could positively regulate STT3A expression through activating c-Jun to bind to STT3A promoter. Functional assays showed that inhibition of TGF-β1 resulted in a decrease of glycosylated PD-L1 and enhanced cytotoxic T-cell function against NPC cells. Analysis of clinical specimens revealed that the expression of STT3A was positively correlated with TGF-β1 and c-Jun, and high STT3A expression was positively correlated with a more advanced clinical stage. Altogether, TGF-β1 activated c-Jun/STT3A signaling pathway to promote N-glycosylation of PD-L1, thus further facilitating immune evasion and reducing the efficacy of cancer immunotherapy. As such, all these data suggested that targeting TGF-β1 pathway might be a promising approach to enhance immune checkpoint blockade, and simultaneous blockade of PD-L1 and TGF-β1 pathways might elicit potent and superior antitumor activity relative to monotherapies.

Keywords: PD-L1; TGF-β1; glycosylation; immunotherapy; nasopharyngeal carcinoma.

PubMed Disclaimer

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Figures

Figure 1
Figure 1
PD-L1 was N-glycosylated in Nasopharyngeal Carcinoma. (A) Western blot analysis of PD-L1 in NPC patient samples. (B) Western blot analysis of PD-L1 in 6 NPC cells including 5-8F, CNE1, CNE2, 6-10B, HNE1, SUNE1. (C) Glycosylation pattern of PD-L1 protein in 5-8F and CNE1 cells. Cell lysates were treated with PNGase F or O-glycosidase and analyzed by western blot analysis. (D) Western blot analysis of PD-L1 in 5-8F and CNE1 cells treated by Tunicamycin (TM) with increasing concentrations.
Figure 2
Figure 2
Inhibition of TGF-β1 by SB431542 Reduced PD-L1 Glycosylation in Vitro. (A) Western blot analysis of PD-L1 in 5-8F and CNE1 cells treated by SB431542 with increasing concentrations. (B) Western blot analysis of PD-L1 and c-Jun in exogenous Flag-PD-L1 expressing 5-8F and CNE1 cells with the treatment of 20μM SB431542. (C) The glycosylation status of PD-L1 protein purified from SB431542 treating cells was analyzed by ConA lectin binding assay.
Figure 3
Figure 3
SB431542 Downregulated Glycosylation of PD-L1 via C-Jun/STT3A Pathway in Vitro and C-Jun was a Direct Transcriptional Regulator of STT3A. (A) Western blot analysis of c-Jun, STT3A and PD-L1 after silencing c-Jun in 5-8F and CNE1 cells. (B) Western blot analysis of STT3A and PD-L1 after silencing STT3A in 5-8F and CNE1 cells. (C) Western blot analysis of c-Jun, STT3A and PD-L1 after overexpressing c-Jun with(out) silencing STT3A in 5-8F and CNE1 cells. (D) Western blot analysis of PD-L1, c-Jun and STT3A in exogenous Flag-PD-L1 expressing 5-8F and CNE1 cells with the treatment of 20μM SB431542. (E) The binding motif of c-Jun from JASPAR database. (F) The CHIP-PCR assay was used to assess the binding of STT3A promoter region. (G) Anti-c-Jun-pulled down chromatins were analyzed by qRT-PCR. (H) A diagram showing the relationship of full-length and mutant STT3A promoters. (I) Dual-luciferase reporter gene assay was performed to indicate the interaction between c-Jun and STT3A. Bars, mean ± SD, *p<0.5, **p<0.01.
Figure 4
Figure 4
STT3A Expression was Positively Correlated with TGF-β1 and C-Jun in NPC Tissues, and High STT3A Expression was Associated with a more advanced stage in NPC. (A) Representative pictures of c-Jun and STT3A expression in NPC patients in TGF-β1-low and high expression groups. Scale bars represented 100μm. (B, C) Pearson correlation analysis of the association between TGF-β1 and STT3A (r=0.412; p=0.013) and between c-Jun and STT3A (r=0.5859; p=0.0002) in NPC tissues. (D) Comparison of STT3A levels in 36 NPC patients with different clinical stages. **p<0.01.
Figure 5
Figure 5
Inhibition of TGF-β1 by SB431542 suppressed T cells activity by inhibiting PD-L1 glycosylation in Vitro. (A, B) The death rate of 5-8F or CNE1 cells co-cultured with Jurkat T cells for 6h. The tumor cells were STT3A-kncokdowned by shRNA or pretreated with SB431542 (20μM) or TM (10μg/ml) for 48h. (C, D) IL-2 production by activated Jurkat T cells co-cultured with 5-8F or CNE1 cells with shSTT3A or pretreated with SB431542 (20μM) or TM (15μg/ml) for 48h. IL-2 secretion was measured by ELISA. Bars, mean ± SD, **p<0.01.

Similar articles

Cited by

References

    1. Chen Y-P, Chan ATC, Le Q-T, Blanchard P, Sun Y, Ma J. Nasopharyngeal Carcinoma. Lancet (2019) 394(10192):64–80. doi: 10.1016/S0140-6736(19)30956-0 - DOI - PubMed
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. . Tumor-Associated B7-H1 Promotes T-Cell Apoptosis: A Potential Mechanism of Immune Evasion. Nat Med (2002) 8(8):793–800. doi: 10.1038/nm730 - DOI - PubMed
    1. Muro K, Chung HC, Shankaran V, Geva R, Catenacci D, Gupta S, et al. . Pembrolizumab for Patients With PD-L1-Positive Advanced Gastric Cancer (KEYNOTE-012): A Multicentre, Open-Label, Phase 1b Trial. Lancet Oncol (2016) 17(6):717–26. doi: 10.1016/S1470-2045(16)00175-3 - DOI - PubMed
    1. Sznol M, Chen L. Antagonist Antibodies to PD-1 and B7-H1 (PD-L1) in the Treatment of Advanced Human Cancer. Clin Cancer Res (2013) 19(5):1021–34. doi: 10.1158/1078-0432.CCR-12-2063 - DOI - PMC - PubMed
    1. Velcheti V, Schalper KA, Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, et al. . Programmed Death Ligand-1 Expression in Non-Small Cell Lung Cancer. Lab Invest (2014) 94(1):107–16. doi: 10.1038/labinvest.2013.130 - DOI - PMC - PubMed