Visual Abstract

Therapies targeting programmed cell death protein 1 (PD-1) have gained great success in patients with multiple types of cancer. The regulatory mechanisms underlying PD-1 expression have been extensively explored. However, the impact of long noncoding RNAs on PD-1 expression remains elusive. In this study, we identified the Notch1/lncNDEPD1 axis, which plays a critical role in PD-1 expression in human CD8+ T cells. RNA sequencing and quantitative reverse transcription PCR data showed that lncNDEPD1 was upregulated in activated T cells, especially in PD-1high subsets. Fluorescence in situ hybridization demonstrated that lncNDEPD1 was localized in the cytoplasm. A mechanistic study showed that lncNDEPD1 could bind with miR-3619-5p and PDCD1 mRNA to prevent PDCD1 mRNA degradation and then upregulate PD-1 expression. A chromatin immunoprecipitation assay showed that Notch1 directly binds to the promoter of lncNDEPD1 instead of PDCD1. Furthermore, chimeric Ag receptor T cells expressing lncNDEPD1-specific short hairpin RNAs were generated. Chimeric Ag receptor T cells with decreased lncNDEPD1 expression showed enhanced tumoricidal effects when PD-L1 was present. Our work uncovered a new regulatory mechanism of PD-1 expression and thus provided a potential target to decrease PD-1 without affecting T cell function.

CD8+ T cells are an essential component of the adaptive immune response that can rapidly expand and differentiate into cytotoxic effector T cells to control and eliminate pathogens upon stimulation. Along with its activation, programmed cell death protein 1 (PD-1) is upregulated and prevents the excessive activation of T cells (1). PD-1 expression is tightly modulated and remains high during persistent stimulation, such as in anti-tumor responses (2, 3). Although PD-1 itself does not impair the effector function of CD8+ T cells, the engagement of programmed cell death protein ligand 1 (PD-L1) with PD-1 can lead to the dysfunction of host cells (4). Hence, T cell–based therapies, including chimeric Ag receptor (CAR)-T cell therapy, have unsatisfactory effects on the treatment of solid tumors (5, 6). To improve the anti-tumor effects of T cells, anti–PD-1 Abs have been used in therapy for patients with solid tumors. Such combinations have yielded satisfactory clinical efficacies (79).

Various factors, including transcription factors (10), epigenetic components (2, 11), and noncoding RNA (ncRNA) (1214), have been reported to affect PD-1 expression. Transcription factors, including Eomes (15), NFATc1 (16), NF-κB (17), T-bet (18), and Notch1 (19, 20), have been well characterized in the regulation of PD-1 expression. Among ncRNAs, microRNAs (miRNAs) have been reported to regulate PD-1 expression through translational repression and mRNA destabilization (21, 22). Long ncRNAs (lncRNAs) also play important roles in regulating gene expression by affecting chromatin modification, transcription, and posttranscriptional processing (2325). It has been reported that lncRNAs are widely expressed in various immune cells and can integrate distinct signal regulators to fine-tune gene expression (26, 27). However, whether lncRNAs regulate PD-1 expression remains unclear.

In this study, we profiled the lncRNAs expressed in activated CD8+ T cells, explored the regulatory roles of lncRNAs on PD-1 expression, and determined the underlying mechanisms of these processes. In addition, we explored the feasibility of lncRNA modification to enhance CAR-T cell function against PD-L1–expressing solid tumors.

Peripheral blood samples from 21 patients with non–small cell lung cancer (NSCLC) were collected from The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Buffy coats from healthy donors were obtained from the Central Blood Bank of Zhengzhou, Zhengzhou, China. The collection and usage of samples from patients and healthy donors were approved by the Ethics Committee of The First Hospital of Zhengzhou University, Zhengzhou, China, and informed consent was obtained from all patients.

PBMCs were isolated through Ficoll-Hypaque density gradient centrifugation as previously described (28). Primary CD8+ T cells were further purified via MACS. The T cells were then activated for 48 h with an activation/expansion kit (Miltenyi Biotec, Bergisch Gladbach, Germany) and IL-2 (100 U/ml, R&D Systems).

After 48 h of activation, PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells were isolated via FACS. Total RNA was extracted using TRIzol reagent (Invitrogen) following the manufacturer’s instructions. Quality control was performed and the certified RNAs were sequenced using a HiSeq PE151 sequencing platform (Illumina).

To detect the competitive interaction between lncNDEPD1 and PDCD1 with miR-3619-5p, we performed a luciferase reporter assay. miRNAs that target both lncNDEPD1 and PDCD1 were predicted using TargetScan (http://www.targetscan.org/) and miRBase (http://www.mirbase.org). The sequences of wild-type (WT) and mutated lncNDEPD1-3′-untranslated region (UTR) and PDCD1-3′-UTR (lncNDEPD1-WT/lncNDEPD1-mutant [MUT], PDCD1-WT/PDCD1-MUT) were synthesized and inserted into the pGL3 luciferase reporter plasmid. Then, WT-3′-UTR or MUT-3′-UTR vectors were cotransfected with the predicted miR-3619-5p mimics or miR-NC into HEK293T cells using Lipofectamine 3000, as described previously (29). Firefly and Renilla luciferase activities were measured using the Dual-Glo luciferase assay system (Promega) 48 h after transfection, and the relative luciferase activity was normalized to Renilla luciferase activity according to the manufacturer’s instructions.

Total RNA was extracted with TRIzol reagent (Invitrogen), and cDNA was prepared using a PrimeScript RT reagent kit with genomic DNA (gDNA) Eraser (Takara) according to the manufacturer’s instructions. Quantitative real-time PCR (qRT-PCR) was performed using SYBR Premix Ex Taq II (Roche) on a Bio-Rad iQ5 iCycler (Bio-Rad). For the reverse transcription of miRNAs, the miScript II RT kit (Qiagen) was used. The relative expression levels of lncRNAs and mRNAs were normalized to that of GAPDH, and miRNAs were normalized to U6 small nuclear RNA. The sequences of the primers used for qRT-PCR were as follows: GAPDH, forward, 5′-ACAACTTTGGTATCGTGGAAGG-3′, reverse, 5′-GCCATCACGCCACAGTTTC-3′; lncNDEPD1, forward, 5′-CGTGATGATGGAGAAGGTCA-3′, reverse, 5′-TACCCATAACGTTGCCCACT-3′; miR-3619-5p, forward, 5′-TCAGCAGGCAGGCTGGTGCAGC-3′, reverse, 5′-CTAGCAAAATAGGCTGTCCC-3′; PDCD1, forward, 5′-TACCCATAACGTTGCCCACT-3′, reverse, 5′-TTTAGCACGAAGCTCTCCGAT-3′.

To determine the presence of surface markers, the cells were washed in FACS buffer (PBS with 2% FBS) and incubated with CD69-FITC (catalog no. 310904, clone FN50, BioLegend), CD8-allophycocyanin-Cy7 (catalog no. 344714, clone SK1, BioLegend), 7-aminoactinomycin D-PerCP (catalog no. 420404, BioLegend), PD-1-PE-Cy7 (catalog no. 329918, clone EH12.2H7, BioLegend), or PD-L1-PE (catalog no. 374512, clone MIH3, BioLegend) Abs for 30 min at 4°C in the dark. The cells were then subjected to FACS analysis. For intracellular staining, CD8+ T cells were stimulated for 6 h with PMA (1 mg/ml, Sigma-Aldrich) and ionomycin (50 ng/ml, Sigma-Aldrich) in the presence of brefeldin A solution (5 mg/ml, BioLegend) at 37°C in a 5% CO2 atmosphere. The cells were fixed, permeabilized, and stained with IL-2-allophycocyanin (catalog no. 500310, clone MQ1-17H12, BioLegend), IFN-γ-allophycocyanin (catalog no. 502512, clone 4S.B3, BioLegend), granzyme B-PE (catalog no. 372208, clone QA16A02, BioLegend) for 30 min at 4°C in the dark. Isotype IgG served as the negative control for cell surface and intracellular staining. Cells were subsequently collected on a BD FACSCanto II flow cytometer (BD Biosciences) and analyzed using FlowJo v7 software (Tree Star).

To detect the expression of lncNDEPD1, fluorescence in situ hybridization (FISH) probes of lncNDEPD1 were designed and synthesized by Ribobio (Guangzhou, China). CD8+ T cells from healthy donors were washed with PBS and fixed in 4% formaldehyde for 10 min on slides. The fixed cells were permeabilized with PBS containing 0.5% Triton X-100 at 4°C for 5 min, washed with PBS three times, and prehybridized at 37°C for 30 min. Hybridization was carried out using anti-lncNDEPD1, anti-18S, and anti-U6 oligodeoxynucleotide probes at 37°C overnight. After incubation, the cells were rinsed with saline-sodium citrate buffer and PBS and were counterstained with DAPI. The images were captured using a fluorescence microscope (IX71, Olympus).

Chromatin immunoprecipitation (ChIP) assays were carried out using the SimpleChIP enzymatic ChIP kit (Cell Signaling Technology) following the manufacturer’s instructions. Briefly, CD8+ T cells were cross-linked using 1% formaldehyde. Then, the cross-linked chromatin DNA was sonicated into 150- to 900-bp fragments. Disrupted chromatin fragments were immunoprecipitated with the corresponding Abs against the protein of interest (anti-Notch1 Ab, Cell Signaling Technology). A positive control (anti-histone H3 Ab) and negative control (anti-IgG Ab) were also detected. The precipitated DNA was purified using spin columns and analyzed via qPCR.

An RNA-binding protein immunoprecipitation (RIP) assay on Ago2 was performed using a Magna RIP kit (Millipore) according to the manufacturer’s instructions. Briefly, CD8+ T cells were lysed using RIP lysis buffer and incubated with anti-Ago2 Abs (Proteintech). The coprecipitated RNAs were isolated and quantified to determine the expression levels of lncNDEPD1, PDCD1, and miR-3619-5p via qRT-PCR. Samples incubated with anti-IgG Ab (Cell Signaling Technology) were used as negative controls. Anti-EZH2 Ab (Millipore) was used as the positive control.

To generate entire DNA sequences of CARs targeting mesothelin (MSLN) and short hairpin RNA (shRNA) targeting lncNDEPD1, the nucleotide sequence of scFv-anti-human MSLN and specific shRNA sequences of lncNDEPD1 were synthesized and fused to the transmembrane (TM) domains of CD28 and CD3ζ. The generated sequence of MSLN-ScFv-Hinge-TM-CD28-CD3ζ, MSLN-shNC-ScFv-Hinge-TM-CD28-CD3ζ, and MSLN-shLNC-ScFv-Hinge-TM-CD28-CD3ζ were subcloned into pCDH-EF1-GFP lentiviral vectors using XhoI and EcoRI restriction sites. Then, the CAR-expressing vectors and packaging vector were transferred into the HEK293T retroviral packaging cells. Lentivirus-containing supernatants were collected after 48–72 h. T cells were transduced with lentiviral particles containing human anti-MSLN scFv (name MSLN-CAR-T) and lncNDEPD1-directed shRNA or a scrambled shRNA control, namely MSLN-CAR-T-SHNC and MSLN-CAR-T-SHLNC, respectively. The transfection efficiency and phenotypic characteristics of CAR-T cells were detected via FACS analysis of GFP expression 72 h later.

The lentiviral vector expressing MSLN-specific CAR and shRNA was constructed as previously reported (11). The shRNA sequences of lncNDEPD1 were as follows: lncNDEPD1 shRNA-1 (target sequence: 5′-GTTCTGTATGAGACCACTTGGATCCGCAAAGTGGGCAACGTTATGAGAATAACGTTGCCCACTTTGCTTTTTGAATTCTTCGATCTGCTTTTT-3′) and lncNDEPD1 shRNA-2 (target sequence: 5′-GTTCTGTATGAGACCACTTGGATCCGCCAGACTCCTGACTAGAAGAGATTCTAGTCAGGAGTCTGGCTTTTTGAATTCTTCGATCTGCTTTTT-3′). The small interfering RNA (siRNA) sequence of Notch1 was as follows: siNotch1 (5′-CAGGGAGCAUGUGUAACAUTTAUGUUACACAUGCUCCCUGTT-3′). T cells were transfected using Lipofectamine 3000 reagent according to the manufacturer’s protocol (Thermo Fisher Scientific, L300015).

CAR-T cells were cocultured with the lung cancer cell line H322 at a 1:1 ratio in a 96-well culture plate. After 24 h of coculture, the release of IFN-γ and IL-2 into the medium was determined using ELISA as described previously (30).

To generate a lung cancer xenograft mouse model, female NOD-SCID mice aged 6–8 wk were purchased from Beijing Vital River Laboratory Animal Technology. FFluc-expressing H322 cells (1 × 106) in 100 μl of PBS were injected s.c. into the mice. Five days later, the tumor-bearing mice were injected once with CAR-T cells or PBS. Tumor growth was monitored using an in vivo imaging system (IVIS Lumina Series III; PerkinElmer) on days 3, 6, 9, and 14 after CAR-T cell infusion. The animal study protocol was approved by the Review Board of The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.

Experiments were performed in triplicate. Statistical analyses were conducted using GraphPad Prism 7 and SPSS Statistics Program (GraphPad Software, La Jolla, CA). The data are presented as the mean ± SEM. Statistical significance was set at p < 0.05. The t test and ANOVA were used to compare the differences among groups. Pearson’s coefficient correlation was used to analyze the correlation between two variables. Statistical significance is indicated as follows: *p < 0.05, ** p < 0.01, ***p < 0.001.

Upon stimulation with anti-CD3/CD28 beads, T cells had upregulated PD-1 expression (Fig. 1A). However, there were fractions of activated T cells (CD69+) that were negative for PD-1 expression (Fig. 1B). PD-1 transcription was confirmed in T cells that were negative for PD-1 or expressed high levels of PD-1. As shown in (Fig. 1C, the mRNA levels of PD-1 were in agreement with the protein production levels, indicating that PD-1 transcription was differentially regulated in activated T cells. In addition to transcription factors, lncRNAs represent a more flexible approach in regulating gene expression. Hence, we analyzed the alterations in lncRNA levels in activated T cells. RNA sequencing data showed that several lncRNAs demonstrated differential expression between T cells expressing high or low levels of PD-1 (Fig. 1D). Among them, NONHSAT068957.2 (hereafter referred to as lncNDEPD1) was confirmed to be overexpressed in PD-1high T cells (Fig. 1E). Further assays showed that lncNDEPD1 was upregulated in active T cells, especially in PD-1high subsets (p < 0.05; (Fig. 1F). FISH assays demonstrated that lncNDEPD1 was mainly located in the cytoplasm of T cells (Fig. 1G). In addition, FISH assays also demonstrated that lncNDEPD1 was gradually upregulated during T cell activation (Fig. 1H). Taken together, these data indicate that lncNDEPD1 is differentially expressed in activated CD8+ T cells with different PD-1 expression levels.

FIGURE 1.

lncNDEPD1 is identified as an upregulated lncRNA correlated with PD-1 expression in activated CD8+ T cells. (A) Representative flow cytometry shows the in vitro expression of PD-1 on CD8+ T cells for the indicated times. (B) Fluorescence cell sorting was performed to obtain PD-1high (top 20%) versus PD-1 (bottom 20%) subsets from CD69+CD8+ T cells. (C) qPCR validation of the differential expression of PDCD1 between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells. Error bars represent the SEM. Data were normalized to GAPDH. (D) Heatmap of differentially expressed lncRNAs in the two cell subsets. (E) Validation of several significantly differentially expressed lncRNAs via qRT-PCR. lncNDEPD1 is identified as an upregulated lncRNA. (F) lncNDEPD1 expression in naive, PD-1high, and PD-1neg populations as analyzed via qRT-PCR. (G) The FISH assay was conducted to confirm that lncNDEPD1 was located mainly in the cytoplasm of activated CD8+ T cells. U6 and 18S were used as controls (representing the nuclear and cytoplasmic fractions, respectively). One representative micrograph is shown (original magnification ×200). (H) A FISH assay was performed to confirm lncNDEPD1 expression. One representative micrograph is shown (original magnification ×200). CD8+ T cells from PBMCs of healthy donors were activated with anti-CD3/CD28 beads for the indicated times to determine lncNDEPD1 expression. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of eight subjects. Data are representative of three independent experiments. **p < 0.01.

FIGURE 1.

lncNDEPD1 is identified as an upregulated lncRNA correlated with PD-1 expression in activated CD8+ T cells. (A) Representative flow cytometry shows the in vitro expression of PD-1 on CD8+ T cells for the indicated times. (B) Fluorescence cell sorting was performed to obtain PD-1high (top 20%) versus PD-1 (bottom 20%) subsets from CD69+CD8+ T cells. (C) qPCR validation of the differential expression of PDCD1 between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells. Error bars represent the SEM. Data were normalized to GAPDH. (D) Heatmap of differentially expressed lncRNAs in the two cell subsets. (E) Validation of several significantly differentially expressed lncRNAs via qRT-PCR. lncNDEPD1 is identified as an upregulated lncRNA. (F) lncNDEPD1 expression in naive, PD-1high, and PD-1neg populations as analyzed via qRT-PCR. (G) The FISH assay was conducted to confirm that lncNDEPD1 was located mainly in the cytoplasm of activated CD8+ T cells. U6 and 18S were used as controls (representing the nuclear and cytoplasmic fractions, respectively). One representative micrograph is shown (original magnification ×200). (H) A FISH assay was performed to confirm lncNDEPD1 expression. One representative micrograph is shown (original magnification ×200). CD8+ T cells from PBMCs of healthy donors were activated with anti-CD3/CD28 beads for the indicated times to determine lncNDEPD1 expression. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of eight subjects. Data are representative of three independent experiments. **p < 0.01.

Close modal

lncRNAs can stabilize mRNAs to promote gene expression. As lncNDEPD1 and PD-1 mRNA showed concerted expression, we speculated whether lncNDEPD1 was involved in PD-1 expression. In CD8+ T cells from patients with NSCLC, the expression of lncNDEPD1 was positively correlated with PD-1 levels (p < 0.0001; (Fig. 2A). Then, T cells were transfected with siRNAs specific for lncNDEPD1 and activated. lncNDEPD1-specific siRNAs markedly downregulated the transcription of lncNDEPD1 and PD-1 (p < 0.01; (Fig. 2B, 2C). In addition, the surface expression of PD-1 protein was downregulated by siRNAs against lncNDEPD1 (p < 0.05; (Fig. 2D, 2E). These results suggest that lncNDEPD1 modulates the mRNA expression of PD-1.

FIGURE 2.

lncNDEPD1 is required for PD-1 expression during CD8+ T cell activation. (A) Scatterplot showing lncNDEPD1 transcript levels from 21 NSCLC samples compared against the transcript levels of PDCD1. The Pearson correlation coefficient is shown. (B) Activated CD8+ T cells were transfected with siRNA targeting lncNDEPD1. qRT-PCR validated the knockdown efficiency of siRNA against lncNDEPD1. (C) qRT-PCR was performed to analyze the alteration of PD-1 expression following knockdown of lncNDEPD1. The statistical data are from three independent experiments, and the bars indicate the SEM. (D) Representative flow cytometric plot of PD-1+CD8+ T cells showing that, compared with siNC, silncNDEPD1 induced significant downregulation of PD-1 expression. (E) Pooled data are presented as the mean ± SEM (n = 3). CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of four subjects. Data are representative of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 2.

lncNDEPD1 is required for PD-1 expression during CD8+ T cell activation. (A) Scatterplot showing lncNDEPD1 transcript levels from 21 NSCLC samples compared against the transcript levels of PDCD1. The Pearson correlation coefficient is shown. (B) Activated CD8+ T cells were transfected with siRNA targeting lncNDEPD1. qRT-PCR validated the knockdown efficiency of siRNA against lncNDEPD1. (C) qRT-PCR was performed to analyze the alteration of PD-1 expression following knockdown of lncNDEPD1. The statistical data are from three independent experiments, and the bars indicate the SEM. (D) Representative flow cytometric plot of PD-1+CD8+ T cells showing that, compared with siNC, silncNDEPD1 induced significant downregulation of PD-1 expression. (E) Pooled data are presented as the mean ± SEM (n = 3). CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of four subjects. Data are representative of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Cytoplasmic lncRNAs have been reported to regulate mRNA levels by acting as decoys to sequester miRNAs (31). Using the TargetScan and miRBase algorithms, five miRNAs were predicted to target both lncNDEPD1 and PD-1. Of these miRNAs, miR-3619-5p was markedly downregulated in PD-1highCD8+ T cell subsets (Fig. 3A). Moreover, miR-3619-5p was increased in T cells transfected with silncNDEPD1 (p < 0.01; (Fig. 3B). Thus, we focused on miR-3619-5p and explored its interaction with lncNDEPD1 and PD-1. Naive CD8+ T cells were transfected with synthesized miR-3619-5p (miR-3619-5p mimics) or a scrambled miRNA (miR-NC as a control). RT-PCR demonstrated that miRNA-3619-5p significantly decreased lncNDEPD1 and PD-1 expression at the mRNA level (p < 0.01; (Fig. 3C). In addition, the surface expression of PD-1 protein was downregulated by miR-3619-5p mimics (p < 0.01; (Fig. 3D, 3E). To further confirm the specific binding of miR-3619-5p to lncNDEPD1 and PDCD1, we performed a RIP assay. The results showed that lncNDEPD1, PDCD1, and miR-3619-5p were predominantly enriched in the group treated with anti-Ago2 Abs (Fig. 3F). As shown in (Fig. 3G, there were complementary sequences among miR-3619-5p, lncNDEPD1, and PDCD1. To test the direct interaction among these three molecules, pmirGLO luciferase reporter vectors expressing lncNDEPD1 with the WT (WT-lncNDEPD1-3′-UTR) or a mutated 3′-UTR (MUT-lncNDEPD1-3′-UTR), and PDCD1 with the WT (WT-PDCD1-3′-UTR) or a MUT 3′-UTR (MUT-PDCD1-3′-UTR) were constructed. These vectors were then cotransfected with miR-3619 or miR-NC into HEK293 cells. miR-3619-5p overexpression reduced the luciferase activity of pmirGLO-lncNDEPD1-WT and pmirGLO-PDCD1-WT (Fig. 3H). In contrast, miR-3619-5p had little impact on the luciferase activity of pmirGLO-lncNDEPD1-MUT and pmirGLO-PDCD1-MUT (p < 0.01; (Fig. 3I). Taken together, these data suggest that lncNDEPD1 can upregulate PD-1 expression by competing with miR-3619-5p.

FIGURE 3.

lncNDEPD1 as competing endogenous RNA competes with PD-1 for the binding of miR-3619-5p. (A) Bioinformatics analysis predicted the miRNAs targeting both lncNDEPD1 and PD-1 in CD8+ T cells and were validated through qRT-PCR. Significantly differentially expressed miR-3619-5p between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells were also identified. (B) qRT-PCR assays determined the expression levels of miR-3619-5p in the silncNDEPD1 or siNC groups. (C) Relative expression of lncNDEPD1 and PD-1 on activated CD8+ T cells transfected with miR-NC or miR-3619-5p mimics were detected using qRT-PCR. (D) Representative flow cytometric plot of PD-1 expression as detected on activated CD8+ T cells after transfection with miR-NC or miR-3619-5p mimics. (E) Graph showing the mean ± SEM of independent experiments. (F) RIP assay showed the enrichment of lncNDEPD1, PD-1, and miR-3619-5p on Ago2 following CD8+ T cell activation. (G) Schematic outline of the predicted binding sites of miR-3619-5p on PDCD1 and lncNDEPD1. Mutations in the binding sites disrupted miR-3619-5p binding capability. (H and I) Luciferase activity was detected in HEK293T cells after transfection with pmiR-GLO vectors containing WT-lncNDEPD1-3′-UTR, MUT-lncNDEPD1-3′-UTR, WT-PDCD1-3′-UTR, and MUT-PDCD1-3′-UTR. Data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of seven subjects. Data are presented as mean ± SEM. Error bars represent SEM from three independent experiments. **p < 0.01, ***p < 0.001.

FIGURE 3.

lncNDEPD1 as competing endogenous RNA competes with PD-1 for the binding of miR-3619-5p. (A) Bioinformatics analysis predicted the miRNAs targeting both lncNDEPD1 and PD-1 in CD8+ T cells and were validated through qRT-PCR. Significantly differentially expressed miR-3619-5p between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells were also identified. (B) qRT-PCR assays determined the expression levels of miR-3619-5p in the silncNDEPD1 or siNC groups. (C) Relative expression of lncNDEPD1 and PD-1 on activated CD8+ T cells transfected with miR-NC or miR-3619-5p mimics were detected using qRT-PCR. (D) Representative flow cytometric plot of PD-1 expression as detected on activated CD8+ T cells after transfection with miR-NC or miR-3619-5p mimics. (E) Graph showing the mean ± SEM of independent experiments. (F) RIP assay showed the enrichment of lncNDEPD1, PD-1, and miR-3619-5p on Ago2 following CD8+ T cell activation. (G) Schematic outline of the predicted binding sites of miR-3619-5p on PDCD1 and lncNDEPD1. Mutations in the binding sites disrupted miR-3619-5p binding capability. (H and I) Luciferase activity was detected in HEK293T cells after transfection with pmiR-GLO vectors containing WT-lncNDEPD1-3′-UTR, MUT-lncNDEPD1-3′-UTR, WT-PDCD1-3′-UTR, and MUT-PDCD1-3′-UTR. Data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of seven subjects. Data are presented as mean ± SEM. Error bars represent SEM from three independent experiments. **p < 0.01, ***p < 0.001.

Close modal

To assess whether lncNDEPD1 and miR-3619-5p affected T cell function, T cells were transfected with siRNAs specific for lncNDEPD1 or miR-3619-5p-mimics when cocultured with A549 cells (non–PD-L1 overexpression). The results showed that lncNDEPD1 knockdown had little effect on T cell function (Fig. 4A). Then, we generated the PD-L1–overexpressing A549 cell lines and detected the overexpression efficiency (Supplemental Fig. 1). We found that knockdown of lncNDEPD1 rescued the inhibition of T cells function by PD-L1, when cocultured with A549 cells with PD-L1 overexpression) (p < 0.05; (Fig. 4B), indicating that lncNDEPD1 affects T cell function by regulating PD-1 expression. Similarly, miR-3619-5p overexpression had little impact on T cell activation in the absence of PD-L1 (Fig. 4C), but remarkably it enhanced the cytotoxic potential of T cells when PD-L1 was present (p < 0.05; (Fig. 4D). These results indicate that miR-3619-5p and lncNDEPD1 affect T cell activity by regulating PD-1/PD-L1 axis–mediated immunosuppression.

FIGURE 4.

lncNDEPD1 and miR-3619-5p impact T cell function in a PD-L1–dependent manner. (A) T cells were transfected with siRNAs specific for lncNDEPD1 and then activated. Representative flow cytometric histograms of functional cytokines including GZMB, IFN-γ, and IL-2 were shown via intracellular cytokine staining in the presence of A549 (non–PD-L1 overexpression). Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (B) T cells were transfected with siRNAs specific for lncNDEPD1, then activated for 48 h in the presence of PD-L1–overexpressing A549 cells. Representative flow cytometric histograms of functional cytokines, including GZMB, IFN-γ, and IL-2, were measured through intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (C) T cells were transfected with miR-3619-5p mimics and miR-NC in the presence of A549 (non–PD-L1 overexpression). Representative flow cytometric histograms showed functional cytokines, including GZMB, IFN-γ, and IL-2, expression on transfected CD8+ T cells through intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (D) T cells were transfected with miR-3619-5p mimics, activated, and then cocultured with A549 cells, which overexpress PD-L1, for 48 h. Representative flow cytometric histograms of functional cytokines, including GZMB, IFN-γ, and IL-2, were measured via intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of six subjects. Data are presented as the mean ± SEM. Error bars represent the SEM from three independent experiments. **p < 0.01, ***p < 0.001.

FIGURE 4.

lncNDEPD1 and miR-3619-5p impact T cell function in a PD-L1–dependent manner. (A) T cells were transfected with siRNAs specific for lncNDEPD1 and then activated. Representative flow cytometric histograms of functional cytokines including GZMB, IFN-γ, and IL-2 were shown via intracellular cytokine staining in the presence of A549 (non–PD-L1 overexpression). Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (B) T cells were transfected with siRNAs specific for lncNDEPD1, then activated for 48 h in the presence of PD-L1–overexpressing A549 cells. Representative flow cytometric histograms of functional cytokines, including GZMB, IFN-γ, and IL-2, were measured through intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (C) T cells were transfected with miR-3619-5p mimics and miR-NC in the presence of A549 (non–PD-L1 overexpression). Representative flow cytometric histograms showed functional cytokines, including GZMB, IFN-γ, and IL-2, expression on transfected CD8+ T cells through intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). (D) T cells were transfected with miR-3619-5p mimics, activated, and then cocultured with A549 cells, which overexpress PD-L1, for 48 h. Representative flow cytometric histograms of functional cytokines, including GZMB, IFN-γ, and IL-2, were measured via intracellular cytokine staining. Pooled data showed the percentage of experimental values relative to the siNC (n = 3). CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of six subjects. Data are presented as the mean ± SEM. Error bars represent the SEM from three independent experiments. **p < 0.01, ***p < 0.001.

Close modal

Notch1 has been reported to regulate PD-1 expression in mouse T cells (20). We wondered whether Notch1 contributes to the differential expression of PD-1 in human T cells. As shown in (Fig. 5A, Notch1 expression was comparable in T cells with different PD-1 expression levels. We knocked down the Notch1 expression using siRNA (Supplemental Fig. 2A). PD-1 was downregulated at both the mRNA and protein levels upon siRNA-mediated knockdown of Notch1 (Fig. 5C–E). The same results were observed using Notch1 inhibitor (Supplemental Fig. 2B). lncNDEPD1 and miR-3619-5p were also altered by Notch1 inhibitor or knockdown (Fig. 5B, 5C). As the potential binding domains of Notch1 existed in the promoters of both PDCD1 and lncNDEPD1 (Fig. 5F, 5G), it was not clear whether Notch1 directly regulated PDCD1 transcription. Therefore, we performed a ChIP assay. The results showed that the regulatory subunits of RBP-Jκ were physically associated with the lncNDEPD1 promoter (p < 0.01; (Fig. 5I) but not with the PDCD1 promoter (p < 0.01; (Fig. 5H), indicating that Notch1 directly regulates lncNDEPD1 expression, which further regulates PD-1 expression by competing for miR-3619-5p.

FIGURE 5.

Notch1 signaling directly regulates lncNDEPD1 expression during CD8+ T cell activation. (A) Intracellular cytokine staining using anti-Notch1 Abs was employed to compare Notch1 expression levels between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells. (B) The Notch1 inhibitor LY3039478 (50 μM) inhibited Notch1 signaling for 48 h, following CD8+ T cell activation. qRT-PCR showed that the Notch1 inhibitor LY3039478 significantly downregulated lncNDEPD1 and PDCD1 expression and upregulated miR-3619-5p expression. (C) qRT-PCR showed that siRNA-mediated knock down of Notch1 significantly downregulated lncNDEPD1 and PDCD1 expression and upregulated miR-3619-5p expression. (D) The representative flow cytometric plot of PD-1+CD8+ T cells showing that a siNotch1 significantly downregulated the expression of PD-1. (E) Pooled data are presented as the mean ± SEM (n = 3), and data are representative of three independent experiments. (F and G) Identification of the potential RBP-Jκ binding sites in the PDCD1 and lncNDEPD1 promoters. Numbers indicate distances (in bp) from the initiation site. (H and I) ChIP of RBP-Jκ at the putative binding sites of PDCD1 and lncNDEPD1 promoters were performed using anti–RBP-Jκ Ab and control Ab IgG. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of six subjects. Error bars represent the SEM from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 5.

Notch1 signaling directly regulates lncNDEPD1 expression during CD8+ T cell activation. (A) Intracellular cytokine staining using anti-Notch1 Abs was employed to compare Notch1 expression levels between PD-1highCD69+CD8+ T cells and PD-1CD69+CD8+ T cells. (B) The Notch1 inhibitor LY3039478 (50 μM) inhibited Notch1 signaling for 48 h, following CD8+ T cell activation. qRT-PCR showed that the Notch1 inhibitor LY3039478 significantly downregulated lncNDEPD1 and PDCD1 expression and upregulated miR-3619-5p expression. (C) qRT-PCR showed that siRNA-mediated knock down of Notch1 significantly downregulated lncNDEPD1 and PDCD1 expression and upregulated miR-3619-5p expression. (D) The representative flow cytometric plot of PD-1+CD8+ T cells showing that a siNotch1 significantly downregulated the expression of PD-1. (E) Pooled data are presented as the mean ± SEM (n = 3), and data are representative of three independent experiments. (F and G) Identification of the potential RBP-Jκ binding sites in the PDCD1 and lncNDEPD1 promoters. Numbers indicate distances (in bp) from the initiation site. (H and I) ChIP of RBP-Jκ at the putative binding sites of PDCD1 and lncNDEPD1 promoters were performed using anti–RBP-Jκ Ab and control Ab IgG. CD8+ T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of six subjects. Error bars represent the SEM from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

To confirm the regulatory effects of lncNDEPD1 on PD-1 expression and T cell function, we employed CAR-T cells for in vitro and in vivo assays. We sorted the CD8+ T cells (Supplemental Fig. 3A) and generated MSLN-directed CAR-T cells expressing scramble or lncNDEPD1-targeted shRNAs (Fig. 6A, 6C). MSLN is a tumor differentiation Ag that was significantly overexpressed in multiple solid tumors and has emerged as an important target of immunotherapies. T cells transduced with lentiviral particles containing human anti-MSLN scFv, named MSLN-CAR-T, were transduced with anti-MSLN scFv and lncNDEPD1-dericted shRNA or a scrambled shRNA control named MSLN-CAR-T-SHNC or MSLN-CAR-T-SHLNC, respectively. To prevent off-target effects, we first examined the MSLN level in CAR-T cells. The results showed no expression of MSLN in CAR-T cells (Supplemental Fig. 3B). Compared with scrambled shRNA-expressing CAR-T cells, shlncNDEPD1-expressing T cells showed markedly reduced PD-1 expression (p < 0.0001; (Fig. 6B). Then, CAR-T cells were coincubated with H322 cells, which expressed high levels of PD-L1 (Supplemental Fig. 4A). In the coincubation assay, shlncNDEPD1 CAR-T cells demonstrated elevated cytotoxicity in H322 cells (Fig. 6D). In the supernatants of CAR-T cells coincubated with tumor cells at a 10:1 ratio, the expression of effector cytokines was increased in the shlncNDEPD1 group (Fig. 6E). Next, we examined the effect of lncNDEPD1 on the anti-tumor effects and PD-1 expression of CAR-T cells. In comparison with the scrambled group, shlncNDEPD1-expressing CAR-T cells demonstrated enhanced tumor inhibition on day 9 (Fig. 6F–H). Additional tumor growth curves (flux values) strengthen the reproducibility of this experiment (Supplemental Fig. 4B). Moreover, shlncNDEPD1-expressing CAR-T cells maintained low levels of lncNDEPD1 in vivo (Fig. 6I). Further analysis showed that shlncNDEPD1-expressing CAR-T cells expanded more efficiently but had lower PD-1 expression (Fig. 6J–L). These data collectively indicate that lncNDEPD1 promotes PD-1 expression and immunosuppression in T cells in vivo.

FIGURE 6.

Adoptive transfer of lncNDEPD1-knockdown CAR-T cells to tumor-bearing NOD-SCID mice suppressed tumor progression. (A) Representative flow cytometric histograms of GFP expression showing the transfection efficiency of MSLN-CAR-T cells, MSLN-CAR-T-SHNC, and MSLN-CAR-T-SHLNC transduced with lentiviral vectors. (B) Representative flow cytometric histograms showing PD-1 expression in the different groups transfected with the corresponding lentiviral vectors. (C) qRT-PCR assays determined the knockdown efficacy of lncNDEPD1. (D) Cytotoxicity of CAR-T cells was assayed via coincubation with H322 cells at E:T ratios ranging from 1:1 to 10:1. (E) Functional cytokines, including IFN-γ, IL-2, and GZMB, secreted by the indicated modified CAR-T cells were measured via ELISA after a 6-h incubation with H322 cells at an E:T ratio of 10:1. Results represent triplicates. Mean ± SEM is shown. (F) Schematic diagram showing the experimental strategy where NOD/SCID mice were s.c. injected with H322 tumor cells with high MSLN and PD-L1 expression followed by adoptive transfer of CAR-T cells or PBS (n = 4 per group). (G) Tumor burden was monitored by measuring luminescence using an IVIS imaging system in the indicated durations after adoptive transfer of CAR-T cells or PBS. (H) Overall kinetics of systemic progression in mice. Each line denotes an individual group (n = 4 per group). (I) qRT-PCR determined the lncNDEPD1 expression levels of CAR-T cells in vivo. (J) The count of CD3+ T cell infiltration per gram of tumor tissues was analyzed after adoptive transfer of CAR-T cells or PBS (n = 4 per group). The mean ± SEM of a representative experiment (n = 4) is shown. (K) Frequency of PD-1+CD3+ T cell populations infiltrating in tumor tissues in mice after adoptive transfer of CAR-T cells or PBS (n = 4 per group). (L) Mean fluorescence intensity (MFI) of PD-1 in CD3+ T cells (n = 4 per group). T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of five subjects. Data are depicted as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 6.

Adoptive transfer of lncNDEPD1-knockdown CAR-T cells to tumor-bearing NOD-SCID mice suppressed tumor progression. (A) Representative flow cytometric histograms of GFP expression showing the transfection efficiency of MSLN-CAR-T cells, MSLN-CAR-T-SHNC, and MSLN-CAR-T-SHLNC transduced with lentiviral vectors. (B) Representative flow cytometric histograms showing PD-1 expression in the different groups transfected with the corresponding lentiviral vectors. (C) qRT-PCR assays determined the knockdown efficacy of lncNDEPD1. (D) Cytotoxicity of CAR-T cells was assayed via coincubation with H322 cells at E:T ratios ranging from 1:1 to 10:1. (E) Functional cytokines, including IFN-γ, IL-2, and GZMB, secreted by the indicated modified CAR-T cells were measured via ELISA after a 6-h incubation with H322 cells at an E:T ratio of 10:1. Results represent triplicates. Mean ± SEM is shown. (F) Schematic diagram showing the experimental strategy where NOD/SCID mice were s.c. injected with H322 tumor cells with high MSLN and PD-L1 expression followed by adoptive transfer of CAR-T cells or PBS (n = 4 per group). (G) Tumor burden was monitored by measuring luminescence using an IVIS imaging system in the indicated durations after adoptive transfer of CAR-T cells or PBS. (H) Overall kinetics of systemic progression in mice. Each line denotes an individual group (n = 4 per group). (I) qRT-PCR determined the lncNDEPD1 expression levels of CAR-T cells in vivo. (J) The count of CD3+ T cell infiltration per gram of tumor tissues was analyzed after adoptive transfer of CAR-T cells or PBS (n = 4 per group). The mean ± SEM of a representative experiment (n = 4) is shown. (K) Frequency of PD-1+CD3+ T cell populations infiltrating in tumor tissues in mice after adoptive transfer of CAR-T cells or PBS (n = 4 per group). (L) Mean fluorescence intensity (MFI) of PD-1 in CD3+ T cells (n = 4 per group). T cells used in the above experiment were all from PBMCs of healthy donors. There were more than three healthy donors in each experiment, and for a total of five subjects. Data are depicted as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

It remains unclear whether lncRNAs regulate PD-1 expression in T cells. In this study, we found that the transcription of lncNDEPD1 was higher in PD-1highCD69+CD8+ T cells than in their PD-1 counterparts. Knockdown of lncNDEPD1 significantly inhibited PD-1 expression (p < 0.001). Moreover, we observed that the blockade of Notch1 signaling could inhibit lncNDEPD1 transcription, further abrogating PD-1 expression. Mechanistically, Notch1 activation directly upregulated lncNDEPD1 transcription by binding to the promoter of lncNDEPD1, which further upregulated PD-1 expression at posttranscriptional levels in vitro and in vivo.

Technological advances in RNA sequencing have shed light on the role of ncRNAs in the immune system. There is growing evidence showing that ncRNAs regulate the differentiation, activation, and proliferation of T cells through transcriptional, posttranscriptional, and epigenetic modification (32, 33). lncRNAs have distinct functions based on their unique subcellular distribution patterns. Cytoplasmic lncRNAs can regulate mRNA levels by acting as decoys to sequester miRNAs and affect posttranscriptional modulation (34). miRNAs are highly conserved small noncoding RNAs that regulate gene expression by binding to the 3′-UTR of their target mRNA (35). Both lncRNAs and miRNAs have been shown to participate in the development and differentiation of T cells (36, 37). Thus, we speculated that ncRNAs regulate PD-1 expression. As expected, we found that a novel lncRNA, lncNDEPD1, could regulate PD-1 expression. lncNDEPD1 was robustly upregulated in the PD-1highCD8+ T cell subset upon activation. Moreover, lncNDEPD1 expression was significantly downregulated along with PD-1 when the Notch signaling pathway was blocked. The results of the ChIP assay showed that Notch1 directly regulated lncNDEPD1 expression instead of PD-1. Thus, we speculated that lncNDEPD1 may be an important regulator of PD-1 expression. Bioinformatics analysis demonstrated that both lncNDEPD1 and PD-1 were targets of miR-3619-5p. Concordantly, we demonstrated that the expression levels of lncNDEPD1 and PD-1 were inversely correlated with miR-3619-5p levels in activated T cells. Further studies showed that miR-3619-5p overexpression reduced the expression of both lncNDEPD1 and PD-1. RIP and luciferase reporter assays further supported that lncNDEPD1 and PD-1 mRNA were able to bind to miR-3619-5p. Taken together, our data suggest that lncNDEPD1 can upregulate PD-1 expression by competitively sponging miR-3619-5p.

The Notch pathway is an evolutionarily conserved signaling pathway that plays a critical role in the development and function of T cells. When activated, cleavage of the Notch intracellular domain (NICD) migrates into the nuclear complex with the transcription factor RBP-Jk. Then, the NICD/RBP-Jk complex upregulates expression of target genes of Notch signaling (38, 39). In human T cells, Notch1 can regulate T cell effector functions at the transcriptional level (40, 41), enhance differentiation toward short-lived effector cells, and maintain memory T cell subsets (42, 43). In mouse T cells, Notch1 upregulates PD-1 expression by directly enhancing PDCD1 transcription (20). However, we noticed that Notch1 does not directly regulate PDCD1 transcription in human T cells. Instead, Notch1 affected PD-1 expression at the posttranscriptional level by modulating lncNDEPD1 expression. As mentioned above, Notch1 is critical for the optimal activation of human T cells. Direct blockade of Notch1 not only inhibits PD-1 expression but also reduces the effector functions of T cells. Therefore, the identification of lncNDEPD1 provides an opportunity to decrease PD-1 expression but not T cell activation. Our data showed that lncNDEPD1 was involved in PD-1 mRNA stabilization but not T cell activation. Targeting lncNDEPD1 can specifically downregulate PD-1 expression without affecting T cell function.

CAR-T cell therapy has emerged as a promising strategy for treating patients with cancer. The effects of PD-1 on CAR-T cells have been investigated using pharmacological agents, Abs, and molecular methods. In our study, knockdown of lncNDEPD1 significantly decreased PD-1 expression and reversed the PD-L1–dependent suppression of T cell function. We generated CAR-T cells expressing anti-lncNDEPD1 shRNAs to knock down lncNDEPD1 expression. In the presence of PD-L1, lncNDEPD1 knockdown enhanced cytokine production and the cytotoxic function of CAR-T cells. Adoptive transfer of lncNDEPD1-knockdown CAR-T cells effectively delayed tumor growth in a mouse model.

In summary, this study demonstrated that Notch1 directly promotes lncNDEPD1 transcription and that lncNDEPD1 upregulates PD-1 expression by sponging miR-3619-5p, which destabilizes PD-1 mRNA. In addition, lncNDEPD1 knockdown can downregulate PD-1 and enhance CAR-T cell therapy efficacy against PD-L1–expressing tumor cells. Our study suggests that lncNDEPD1 may be a potential target for decreasing PD-1 expression in T cells.

We are grateful for the sample collection performed by the cancer center at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Also, we thank Editage for English language editing.

This work was supported by Natural Science Foundation of China Major Research Plan Grant 91942314, Ministry of Science and Technology of China National Science and Technology Infrastructure Program (National Key Science Projects Program) Grant 2020ZX09201-009, and Natural Science Foundation of China (Foundation for Innovative Research Groups of the National Natural Science Foundation of China) Grant 81771781.

The online version of this article contains supplemental material.

Abbreviations used in this article:

CAR-T

chimeric Ag receptor T

ChIP

chromatin immunoprecipitation

FISH

fluorescence in situ hybridization

lncRNA

long ncRNA

MSLN

mesothelin

miRNA

microRNA

MUT

mutant

ncRNA

noncoding RNA

NSCLC

non–small cell lung cancer

PD-1

programmed cell death protein 1

PD-L1

programmed cell death protein ligand 1

qRT-PCR

quantitative real-time PCR

RIP

RNA-binding protein immunoprecipitation

shRNA

short hairpin RNA

siRNA

small interfering RNA

TM

transmembrane

UTR

untranslated region

WT

wild-type

1.
Wei
F.
,
S.
Zhong
,
Z.
Ma
,
H.
Kong
,
A.
Medvec
,
R.
Ahmed
,
G. J.
Freeman
,
M.
Krogsgaard
,
J. L.
Riley
.
2013
.
Strength of PD-1 signaling differentially affects T-cell effector functions.
Proc. Natl. Acad. Sci. USA
110
:
E2480
E2489
.
2.
Youngblood
B.
,
K. J.
Oestreich
,
S. J.
Ha
,
J.
Duraiswamy
,
R. S.
Akondy
,
E. E.
West
,
Z.
Wei
,
P.
Lu
,
J. W.
Austin
,
J. L.
Riley
, et al
2011
.
Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells.
Immunity
35
:
400
412
.
3.
Xiao
G.
,
A.
Deng
,
H.
Liu
,
G.
Ge
,
X.
Liu
.
2012
.
Activator protein 1 suppresses antitumor T-cell function via the induction of programmed death 1.
Proc. Natl. Acad. Sci. USA
109
:
15419
15424
.
4.
Goods
B. A.
,
A. L.
Hernandez
,
D. E.
Lowther
,
L. E.
Lucca
,
B. A.
Lerner
,
M.
Gunel
,
K.
Raddassi
,
V.
Coric
,
D. A.
Hafler
,
J. C.
Love
.
2017
.
Functional differences between PD-1+ and PD-1 CD4+ effector T cells in healthy donors and patients with glioblastoma multiforme.
PLoS One
12
:
e0181538
.
5.
D’Aloia
M. M.
,
I. G.
Zizzari
,
B.
Sacchetti
,
L.
Pierelli
,
M.
Alimandi
.
2018
.
CAR-T cells: the long and winding road to solid tumors.
Cell Death Dis.
9
:
282
.
6.
Zolov
S. N.
,
S. P.
Rietberg
,
C. L.
Bonifant
.
2018
.
Programmed cell death protein 1 activation preferentially inhibits CD28.CAR-T cells.
Cytotherapy
20
:
1259
1266
.
7.
Rafiq
S.
,
O. O.
Yeku
,
H. J.
Jackson
,
T. J.
Purdon
,
D. G.
van Leeuwen
,
D. J.
Drakes
,
M.
Song
,
M. M.
Miele
,
Z.
Li
,
P.
Wang
, et al
2018
.
Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo.
Nat. Biotechnol.
36
:
847
856
.
8.
Fedorov
V. D.
,
M.
Themeli
,
M.
Sadelain
.
2013
.
PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses.
Sci. Transl. Med.
5
:
215ra172
.
9.
Tian
Y.
,
Y.
Li
,
Y.
Shao
,
Y.
Zhang
.
2020
.
Gene modification strategies for next-generation CAR T cells against solid cancers.
J. Hematol. Oncol.
13
:
54
.
10.
Bally
A. P.
,
J. W.
Austin
,
J. M.
Boss
.
2016
.
Genetic and epigenetic regulation of PD-1 expression.
J. Immunol.
196
:
2431
2437
.
11.
Shi
X.
,
D.
Zhang
,
F.
Li
,
Z.
Zhang
,
S.
Wang
,
Y.
Xuan
,
Y.
Ping
,
Y.
Zhang
.
2019
.
Targeting glycosylation of PD-1 to enhance CAR-T cell cytotoxicity.
J. Hematol. Oncol.
12
:
127
.
12.
Xu
S.
,
Z.
Tao
,
B.
Hai
,
H.
Liang
,
Y.
Shi
,
T.
Wang
,
W.
Song
,
Y.
Chen
,
J.
OuYang
,
J.
Chen
, et al
2016
.
miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint.
Nat. Commun.
7
:
11406
.
13.
Mastroianni
J.
,
N.
Stickel
,
H.
Andrlova
,
K.
Hanke
,
W.
Melchinger
,
S.
Duquesne
,
D.
Schmidt
,
M.
Falk
,
G.
Andrieux
,
D.
Pfeifer
, et al
2019
.
miR-146a controls immune response in the melanoma microenvironment.
Cancer Res.
79
:
183
195
.
14.
Zhao
L.
,
Y.
Liu
,
J.
Zhang
,
Y.
Liu
,
Q.
Qi
.
2019
.
lncRNA SNHG14/miR-5590-3p/ZEB1 positive feedback loop promoted diffuse large B cell lymphoma progression and immune evasion through regulating PD-1/PD-L1 checkpoint.
Cell Death Dis.
10
:
731
.
15.
Zhang
Z.
,
F.
Li
,
Y.
Tian
,
L.
Cao
,
Q.
Gao
,
C.
Zhang
,
K.
Zhang
,
C.
Shen
,
Y.
Ping
,
N. R.
Maimela
,
L.
Wang
,
B.
Zhang
,
Y.
Zhang
.
2020
.
Metformin enhances the antitumor activity of CD8+ T lymphocytes via the AMPK-miR-107-Eomes-PD-1 pathway.
J. Immunol.
204
:
2575
2588
.
16.
Oestreich
K. J.
,
H.
Yoon
,
R.
Ahmed
,
J. M.
Boss
.
2008
.
NFATc1 regulates PD-1 expression upon T cell activation.
J. Immunol.
181
:
4832
4839
.
17.
Bally
A. P.
,
P.
Lu
,
Y.
Tang
,
J. W.
Austin
,
C. D.
Scharer
,
R.
Ahmed
,
J. M.
Boss
.
2015
.
NF-κB regulates PD-1 expression in macrophages.
J. Immunol.
194
:
4545
4554
.
18.
Kao
C.
,
K. J.
Oestreich
,
M. A.
Paley
,
A.
Crawford
,
J. M.
Angelosanto
,
M. A.
Ali
,
A. M.
Intlekofer
,
J. M.
Boss
,
S. L.
Reiner
,
A. S.
Weinmann
,
E. J.
Wherry
.
2011
.
Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection.
Nat. Immunol.
12
:
663
671
.
19.
Pan
T.
,
Z.
Liu
,
J.
Yin
,
T.
Zhou
,
J.
Liu
,
H.
Qu
.
2015
.
Notch signaling pathway was involved in regulating programmed cell death 1 expression during sepsis-induced immunosuppression.
Mediators Inflamm.
2015
:
539841
.
20.
Mathieu
M.
,
N.
Cotta-Grand
,
J. F.
Daudelin
,
P.
Thébault
,
N.
Labrecque
.
2013
.
Notch signaling regulates PD-1 expression during CD8+ T-cell activation.
Immunol. Cell Biol.
91
:
82
88
.
21.
Boldrini
L.
,
M.
Giordano
,
C.
Niccoli
,
F.
Melfi
,
M.
Lucchi
,
A.
Mussi
,
G.
Fontanini
.
2017
.
Role of microRNA-33a in regulating the expression of PD-1 in lung adenocarcinoma.
Cancer Cell Int.
17
:
105
.
22.
Moffett
H. F.
,
A. N. R.
Cartwright
,
H. J.
Kim
,
J.
Godec
,
J.
Pyrdol
,
T.
Äijö
,
G. J.
Martinez
,
A.
Rao
,
J.
Lu
,
T. R.
Golub
, et al
2017
.
The microRNA miR-31 inhibits CD8+ T cell function in chronic viral infection. [Published erratum appears in 2017 Nat. Immunol. 18: 1173.]
Nat. Immunol.
18
:
791
799
.
23.
Wang
K. C.
,
H. Y.
Chang
.
2011
.
Molecular mechanisms of long noncoding RNAs.
Mol. Cell
43
:
904
914
.
24.
Schmitt
A. M.
,
H. Y.
Chang
.
2016
.
Long noncoding RNAs in cancer pathways.
Cancer Cell
29
:
452
463
.
25.
Flynn
R. A.
,
H. Y.
Chang
.
2014
.
Long noncoding RNAs in cell-fate programming and reprogramming.
Cell Stem Cell
14
:
752
761
.
26.
Turner
M.
,
A.
Galloway
,
E.
Vigorito
.
2014
.
Noncoding RNA and its associated proteins as regulatory elements of the immune system.
Nat. Immunol.
15
:
484
491
.
27.
Atianand
M. K.
,
D. R.
Caffrey
,
K. A.
Fitzgerald
.
2017
.
Immunobiology of long noncoding RNAs.
Annu. Rev. Immunol.
35
:
177
198
.
28.
Liu
J. Y.
,
F.
Li
,
L. P.
Wang
,
X. F.
Chen
,
D.
Wang
,
L.
Cao
,
Y.
Ping
,
S.
Zhao
,
B.
Li
,
S. H.
Thorne
, et al
2015
.
CTL- vs Treg lymphocyte-attracting chemokines, CCL4 and CCL20, are strong reciprocal predictive markers for survival of patients with oesophageal squamous cell carcinoma.
Br. J. Cancer
113
:
747
755
.
29.
Cao
L.
,
Y.
Liu
,
D.
Wang
,
L.
Huang
,
F.
Li
,
J.
Liu
,
C.
Zhang
,
Z.
Shen
,
Q.
Gao
,
W.
Yuan
,
Y.
Zhang
.
2018
.
miR-760 suppresses human colorectal cancer growth by targeting BATF3/AP-1/cyclinD1 signaling.
J. Exp. Clin. Cancer Res.
37
:
83
.
30.
Li
F.
,
Z.
Zhang
,
Y.
Xuan
,
D.
Zhang
,
J.
Liu
,
A.
Li
,
S.
Wang
,
T.
Li
,
X.
Shi
,
Y.
Zhang
.
2020
.
PD-1 abrogates the prolonged persistence of CD8+ CAR-T cells with 4-1BB co-stimulation.
Signal Transduct. Target. Ther.
5
:
164
.
31.
Noh
J. H.
,
K. M.
Kim
,
W. G.
McClusky
,
K.
Abdelmohsen
,
M.
Gorospe
.
2018
.
Cytoplasmic functions of long noncoding RNAs.
Wiley Interdiscip. Rev. RNA
9
:
e1471
.
32.
Chen
Y. G.
,
A. T.
Satpathy
,
H. Y.
Chang
.
2017
.
Gene regulation in the immune system by long noncoding RNAs.
Nat. Immunol.
18
:
962
972
.
33.
Mowel
W. K.
,
J. J.
Kotzin
,
S. J.
McCright
,
V. D.
Neal
,
J.
Henao-Mejia
.
2018
.
Control of immune cell homeostasis and function by lncRNAs.
Trends Immunol.
39
:
55
69
.
34.
Chen
L. L.
2016
.
Linking long noncoding RNA localization and function.
Trends Biochem. Sci.
41
:
761
772
.
35.
Krol
J.
,
I.
Loedige
,
W.
Filipowicz
.
2010
.
The widespread regulation of microRNA biogenesis, function and decay.
Nat. Rev. Genet.
11
:
597
610
.
36.
Rossi
R. L.
,
G.
Rossetti
,
L.
Wenandy
,
S.
Curti
,
A.
Ripamonti
,
R. J.
Bonnal
,
R. S.
Birolo
,
M.
Moro
,
M. C.
Crosti
,
P.
Gruarin
, et al
2011
.
Distinct microRNA signatures in human lymphocyte subsets and enforcement of the naive state in CD4+ T cells by the microRNA miR-125b.
Nat. Immunol.
12
:
796
803
.
37.
Hu
G.
,
Q.
Tang
,
S.
Sharma
,
F.
Yu
,
T. M.
Escobar
,
S. A.
Muljo
,
J.
Zhu
,
K.
Zhao
.
2013
.
Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation.
Nat. Immunol.
14
:
1190
1198
.
38.
Kelliher
M. A.
,
J. E.
Roderick
.
2018
.
NOTCH signaling in T-cell-mediated anti-tumor immunity and t-cell-based immunotherapies.
Front. Immunol.
9
:
1718
.
39.
Kondo
T.
,
R.
Morita
,
Y.
Okuzono
,
H.
Nakatsukasa
,
T.
Sekiya
,
S.
Chikuma
,
T.
Shichita
,
M.
Kanamori
,
M.
Kubo
,
K.
Koga
, et al
2017
.
Notch-mediated conversion of activated T cells into stem cell memory-like T cells for adoptive immunotherapy.
Nat. Commun.
8
:
15338
.
40.
Kuijk
L. M.
,
M. I.
Verstege
,
N. V.
Rekers
,
S. C.
Bruijns
,
E.
Hooijberg
,
B. O.
Roep
,
T. D.
de Gruijl
,
Y.
van Kooyk
,
W. W.
Unger
.
2013
.
Notch controls generation and function of human effector CD8+ T cells.
Blood
121
:
2638
2646
.
41.
Cho
O. H.
,
H. M.
Shin
,
L.
Miele
,
T. E.
Golde
,
A.
Fauq
,
L. M.
Minter
,
B. A.
Osborne
.
2009
.
Notch regulates cytolytic effector function in CD8+ T cells.
J. Immunol.
182
:
3380
3389
.
42.
Mathieu
M.
,
F.
Duval
,
J. F.
Daudelin
,
N.
Labrecque
.
2015
.
The Notch signaling pathway controls short-lived effector CD8+ T cell differentiation but is dispensable for memory generation.
J. Immunol.
194
:
5654
5662
.
43.
Tsukumo
S. I.
,
K.
Yasutomo
.
2018
.
Regulation of CD8+ T cells and antitumor immunity by Notch signaling.
Front. Immunol.
9
:
101
.

The authors have no financial conflicts of interest.

Supplementary data