Tumor-specific CD8+ T cells play a pivotal role in antitumor immunity and are a key target of immunotherapeutic approaches. Intratumoral CD8+ T cells are heterogeneous; Tcf1+ stemlike CD8+ T cells give rise to their cytotoxic progeny—Tim-3+ terminally differentiated CD8+ T cells. However, where and how this differentiation process occurs has not been elucidated. We herein show that terminally differentiated CD8+ T cells can be generated within tumor-draining lymph nodes (TDLN) and that CD69 expression on tumor-specific CD8+ T cells controls its differentiation process through regulating the expression of the transcription factor TOX. In TDLNs, CD69 deficiency diminished TOX expression in tumor-specific CD8+ T cells, and consequently promoted generation of functional terminally differentiated CD8+ T cells. Anti-CD69 administration promoted the generation of terminally differentiated CD8+ T cells, and the combined use of anti-CD69 and anti–programmed cell death protein 1 (PD-1) showed an efficient antitumor effect. Thus, CD69 is an attractive target for cancer immunotherapy that synergizes with immune checkpoint blockade.

There are two major subpopulations of intratumoral CD8+ T cells in human and murine cancers (1–3). One is noncytotoxic, stemlike PD-1+Tcf1+Tim-3 CD8+ T-cells that persist long-term and importantly serve as a source of the other subpopulation, terminally differentiated PD-1+Tcf1Tim-3+ CD8+ T cells, which have reduced longevity. Because terminally differentiated CD8+ T cells are the primary cytotoxic populations in the tumor microenvironment (TME), the efficient generation of terminally differentiated CD8+ T cells is critical for antitumor immune responses (4, 5). Recent reports have shown that tumor-draining lymph nodes (TDLN), initial sites of the T-cell immune response, are enriched with tumor-specific stemlike CD8+ T cells and are crucial for the antitumor effect of anti–programmed cell death protein 1 (PD-1)/ programmed death ligand 1 (PD-L1) blockade (6–9), raising the importance of TDLNs for an efficient antitumor immune response. However, the molecular mechanisms underlying how and where tumor-specific stemlike CD8+ T cells give rise to terminally differentiated CD8+ T cells has not been elucidated.

CD69, a type-2 transmembrane glycoprotein, has been known as an activation marker on leukocytes, and recent studies have shown that CD69 is a functional molecule rather than a simple activation marker (10). CD69 suppresses the function of S1P1, thereby inhibiting lymphocyte egress from lymphoid organs (10–12). Furthermore, CD69 interacts with myosin light chain 9/12 (Myl9/12), playing a role in recruiting T cells to inflamed tissues (13). CD69 is also known to be expressed on CD8+ T cells in TDLNs and the TME in human patients with cancer, suggesting its role in antitumor immunity (14). Indeed, we and other groups have independently demonstrated that anti-CD69 treatment enhances antitumor immunity, with similar effects observed in CD69-deficient mice (15–18). These data suggest that CD69 negatively regulates antitumor immunity; however, the detailed molecular mechanisms of CD69-mediated regulation of antitumor immunity have not been elucidated. In this study, our single-cell transcriptomics defined CD69 as an important regulator for differentiation of tumor-specific CD8+ T cells. CD69 deficiency resulted in decreased expression of the transcription factor TOX in tumor-specific CD8+ T cells in TDLNs, promoting differentiation of stemlike CD8+ T cells into the functional terminally differentiated CD8+ T cells in TDLNs. Our results suggest that CD69 imposes tumor-specific CD8+ T-cell fate within TDLNs.

Mice

BALB/c mice and C57BL/6 mice were purchased from Charles River Laboratories Japan or CLEA Japan, respectively. OT-I transgenic mice expressing TCR specific for H-2Kb/OVA257−264 was purchased from The Jackson Laboratory. CD69-deficient mice were established in our laboratory (19) and backcrossed more than 15 times. All mice were maintained under specific pathogen-free conditions and used at 6 to 16 weeks of age. All animal experiments were approved by the Chiba University Review Board for Animal Care.

Cell lines

The murine CT26 colon carcinoma cell line was purchased from ATCC. The murine MC38 colon carcinoma cell line was purchased from Kerafast. Both cell lines have been verified by short tandem repeat (STR) analysis by the providers and frozen in multiple cryovials immediately upon receipt. MC38-OVA cells were made in our laboratory. In brief, chicken OVA cDNA was retrovirally introduced into MC38 cells using pMSCV-OVA-IRES-EGFP vector (VectorBuilder). The murine B16 melanoma expressing chicken OVA was kindly provided by Dr. Nakatsura (National Cancer Center, Tokyo, Japan). CT26 cells were cultured in RPMI 1640 (Sigma) supplemented with 10% FBS (Hyclone), and MC38-OVA cells and B16-OVA cells were cultured in DMEM (Thermo Fisher Scientific) supplemented with 10% FBS. After thawing, cells were passaged a minimum of three times before use in experiments and maintained for no longer than 3 months. All cells were regularly tested for Mycoplasma contamination by PCR.

Tumor model

CT26 (1 × 106) cells were subcutaneously inoculated into the left flank of BALB/c mice. MC38-OVA (1 × 106), and B16-OVA (5 × 105) were subcutaneously inoculated into the left flank of C57BL/6 mice. Tumor growth was monitored by periodic caliper measurements, as previously described (20). FTY720 (20 μg; Cayman chemical) was intraperitoneally administered every other day. Monoclonal anti-mouse CD69 (H1.2F3), anti-mouse PD-1 (RMP1–14, Bio X Cell), or isotype control antibody (each 200 μg) were intraperitoneally administered twice a week.

T-cell isolation

Tumor tissues from all tumor models were minced into small pieces and dissociated using a mouse Tumor Dissociation Kit (Miltenyi Biotec) and Gentle MACS Octo Dissociator with Heaters (Miltenyi Biotec), as described in the manufacturer's protocol. Dissociated cells were filtered through a 70-μm mesh and separated by Percoll (Cytiva) density gradient centrifugation to enrich tumor-infiltrating lymphocytes. ACK lysis buffer (Thermo Fisher Scientific) was used for lysis of red blood cells. Cells from lymph nodes were isolated by gently teasing between frosted glass slides and filtered through a 70-μm mesh. Because tumor cells were subcutaneously inoculated into the left flank of mice, the left axillary lymph node was considered as the TDLN and the right axillary lymph node was considered as the non-TDLN. Naïve CD8+ T cells were isolated from spleen by negative selection using MojoSort Mouse CD8 Naive T-cell Isolation Kit (BioLegend) with MACS column (Miltenyi Biotec). More than 95% of the isolated cells were CD62L+CD44lo CD8+ T cells, determined by flow cytometry.

Flow cytometry

CD8+ T cells or DCs in TDLN or TME were stained with following fluorochrome-conjugated monoclonal antibodies. CD8α (53–6.7), CD11b (M1/70), CD11c (N418), CD24 (M1/69), CD44 (IM7), CD45.2 (104), CD62 L (MEL-14), CD69 (H1.2F3), CD80 (16–10A1), CD86 (GL1), CD94 (18d3), Lag-3 (C9B7W), Ly6c (HK1.4), Ly108 (330-AJ), MHC Class II (I-A/I-E) (M5/114.15.2), NFAT2 (7A6), PD-1 (29F.1A12), Tigit (1G9), Tim-3 (RMT3–23) were obtained from Biolegend; CD45 (30–F11), CD45.1 (A20) were obtained from BD Biosciences; Eomes (Dan11mag), Granzyme B (GB11) were obtained from Thermo Fisher Scientific; Tcf1 (C63D9) was obtained from Cell Signaling Technology; TOX (REA473) was obtained from Miltenyi Biotec. Propidium Iodide (PI; Fujifilm) or Fixable Viability Dye eFluor 506 (Thermo Fisher Scientific) were used to exclude dead cells.

For intracellular staining, cells were fixed/permeabilized with the FOXP3 Staining Buffer Set (Thermo Fisher Scientific) according to the manufacturer's instructions. AH1-specific CD8+ T cells were detected with an APC-conjugated MuLV gp70423–431 H-2Ld tetramer (NIH Tetramer Core Facility), and OVA-specific CD8+ T cells were detected with an APC-conjugated chicken OVA257–264 (SIINFEKL) H-2Kb tetramer (NIH Tetramer Core Facility). The following gating strategies were used: Tcf1+ stemlike CD8+ T cells (CD8α+tetramer+PD-1+Tcf1+Tim-3); Tim-3+ terminally differentiated CD8+ T cells (CD8α+tetramer+PD-1+Tcf1Tim-3+). Data were acquired on a BD FACSCanto II flow cytometer (BD Biosciencies) or LSRFortessa flow cytometer (BD Biosciences) and analyzed with the FlowJo software program (BD Biosciences). For live cell sorting of Tcf1+ stemlike or Tim-3+ terminally differentiated CD8+ T cells, the cell surface marker Ly108 was used instead of Tcf1 (1). A FACSAria III cell sorter (BD Biosciences) was used for live cell sorting.

Adoptive cell transfer

Naïve CD8+ T cells were isolated from spleen of CD69-sufficient (CD45.2+) or -deficient (CD45.1+) OT-I mice. An equal mixture of naïve OT-I CD8+ T cells (5×104 each) were intravenously injected into the lateral tail vein of C57BL/6 mice (CD45.1+/CD45.2+). MC38-OVA cells were subcutaneously inoculated on the next day, and OT-I CD8+ T cells in TDLN were analyzed by flow cytometry.

Flow cytometry analysis of nuclear NFAT2

CD8α+PD-1+ cells were sorted from TDLNs of CT26-bearing BALB/c mice using a BD FACS Aria III. Nuclei of T cells were isolated using a protocol adapted from a previous report (21). Briefly, cells were resuspended in buffer containing 320 mmol/L sucrose, 10 mmol/L HEPES (Thermo Fisher Scientific), 8 mmol/L MgCl2, and 0.1% (vol/vol) Triton X-100 (Sigma) for 10 minutes. The lysed cells were centrifuged to collect the nuclei, and the nuclear pellet was fixed in 3% paraformaldehyde (PFA) for 15 minutes. The fixed nuclei were permeabilized with 0.3% Triton X-100 in PBS plus 2% FBS for 10 minutes and stained with a fluorochrome-conjugated anti-NFAT2 (7A6; Biolegend). The nuclei were counterstained with To-Pro-3 (Thermo Fisher Scientific).

T-cell culture

TDLN cells were isolated from CT26-bearing BALB/c mice and cultured in RPMI1640 supplemented with 10% FBS, 5 ng/mL human IL2 (Peprotech), 10 mmol/L HEPES, 1 mmol/L sodium pyruvate (Thermo Fisher Scientific), 1% MEM nonessential amino acids (Thermo Fisher Scientific), 55 μmol/L 2-mercaptoethanol (Thermo Fisher Scientific) in the presence or absence of the calcineurin inhibitor cyclosporin A (10 nmol/L; Nacalai) for 4 hours. CD8α+AH1 tetramer+ cells in TDLNs were analyzed by flow cytometry.

Single-cell RNA sequencing

CD8α+AH1 tetramer+ cells sorted from TDLN or TME of CT26-bearing BALB/c mice using a BD FACS Aria III (10,000 cells each) were encapsulated into droplets, and libraries were prepared using Chromium Single-Cell 3′ Reagent Kits v3.1 according to manufacturer's protocol (10× Genomics). The generated single-cell RNA sequencing (scRNA-seq) libraries were sequenced on a NovaSeq 6000 (Illumina) with 28 cycles for Read 1, 8 cycles for i7 index, and 91 cycles for Read 2. Alternatively, 50 ng of scRNA-seq libraries were converted into a compatible library with MGI sequencer using the MGIEasy Universal Library Conversion kit (MGI). The converted libraries were sequenced on a DNBSEQ-400RS (MGI) with 28 cycles for Read 1, 8 cycles for i7 index, and 91 cycles for Read 2.

Analysis and visualization of scRNA-seq data

Sequence reads from all samples were processed and aggregated using Cell Ranger v.6.0.0 (10× Genomics) based on mouse genome assembly mm10. Aggregated data were further analyzed using Seurat v.4.0.1 (22). Specifically, the expression matrix was log-normalized, and the data were regressed against the total number of unique molecular identifiers (UMI) detected per cell. Principal component analysis (PCA) was performed, and PCA dimensions 1 to 50 were used to find clusters. Finally, marker genes for these clusters were identified. Clusters expressing two or more canonical cell-type markers were classified as doublet cells and excluded from further analyses. The single-cell gene expression was visualized as Uniform Manifold Approximation and Projection (UMAP) overlays, marker gene heat maps, and violin plots using Seurat. The cell-cycle phase score was calculated using Seurat based on the gene list described in Tirosh and colleagues (23). The single-cell gene expression was also visualized as density plots using the Nebulosa R software package v.1.0.1 algorithm (24). The activated pathways were detected by a single-sample gene set variation analysis (ssGSVA; ref. 25). The pathway genes of CA-RIT-NFAT1_up and CA-RIT-NFAT1_down were obtained from (GSE64409; ref. 26). The top 500 differentially expressed genes were determined. The ssGSVA scores were visualized with UMAP overlays and box plots using ggplot2 v.3.3.5.

Bulk RNA-seq

RNA-seq was carried out as previously described (27). Total cellular RNA of stemlike CD8+ T cells (CD8α+AH1 tetramer+PD-1+Ly108+Tim-3) or terminally differentiated CD8+ T cells (CD8α+AH1 tetramer+PD-1+Ly108Tim-3+) sorted from TME of CT26-bearing BALB/c mice was extracted with TRIzol reagent (Thermo Fisher Scientific). For cDNA library construction, a SMARTer Stranded Total RNA-Seq Kit v2 - Pico Input Mammalian (Takara) was used in accordance with the manufacturer's protocol. 20 ng total RNA was used for each sample. Sequencing of library fragments was performed with 76 cycles for Read 1, 8 cycles for index1, and 8 cycles for index2 using an Illumina HiSeq 1500 System. Bcl2fastq v.2.20 was used to convert sequence data to fastq format. FastQC v.0.1.18 was used to check the quality of sequence. For the data analyses, read sequences (50 bp) were aligned to the mm10 mouse reference genome (University of California, Santa Cruz, December 2011) using Bowtie v.2.3.4 and Hisat v.2.1.0. Fragments per kilobase of exon per million mapped reads for each gene were calculated using Cufflinks v.2.2.1, Picard v.2.18.21 and Samtools v.1.9 were used as command line tools to manipulate the SAM/BAM format.

The analysis and visualization of RNA-seq data

Genes in which maximum FPKM value of each gene was ≥ 1.0 were used for further analyses. Gene clustering was performed on the basis of Pearson correlation coefficient using APCluster (an R Package for Affinity Propagation Clustering). PCA was perform using R. A GSEA was performed using the Python package (28). The MA plots depicts was constructed using the ggplot2 v.3.3.5.

Comparison of RNA-seq data

To compare our data with other RNA-seq datasets from the relevant literature, the top 250 differentially expressed genes were determined between the two indicated states for each generated sample. The gene sets of B16-OVA melanoma stemlike and B16-OVA melanoma terminal differentiation were determined using the gene expression profiles of CD8+ T-cell subpopulations in B16-OVA melanoma-bearing mice (GSE122713; ref. 1). The gene sets of LCMV C13 stemlike and LCMV C13 terminal differentiation were determined using the gene expression profiles of CD8+ T-cell subpopulations in LCMV clone-13–infected mice (GSE122713). The gene sets of ToxWT LCMV C13 Tim-3 (stemlike) and ToxKO LCMV C13 Tim-3 (stemlike) were determined using the gene expression profiles of Tim-3 CD8+ T cells in LCMV clone-13–infected mice (GSE131643; ref. 29). The gene sets of ToxWT LCMV C13 Tim-3+ (term diff) and ToxKO LCMV C13 Tim-3+ (term diff) were determined using the gene expression profiles of Tim-3+ CD8+ T cells in LCMV clone-13-infected mice (GSE131643).

qRT-PCR

Total cellular RNA was extracted with TRIzol reagent, and cDNA was synthesized with ReverTra Ace qPCR RT Master Mix (Toyobo). 10 ng total RNA was used for each sample. Quantitative RT-PCR was performed with a StepOnePlus Real-Time PCR System (Thermo Fisher Scientific) and TaqMan Fast Advanced Master Mix (Thermo Fisher Scientific). The TaqMan probes were purchased from Roche. Relative gene expression was calculated from CT values using a ΔΔCt method. The expression of the target genes was normalized to the expression of Actb. The following probes and primers were used to measure the expression of each gene. Actb, probe (#64, catalog no. 04688635001); forward primer 5′-CTAAGGCCAACCGTGAAAAG-3′; and reverse primer 5′-ACCAGAGGCATACAGGGACA-3′. Tox, probe (#42, catalog no. 04685016001); forward primer 5′-GCAAGTCTGCAACCCCTTC-3′; and reverse primer 5′-CGCTTCTCTCCTCCATTGAT-3′.

Dataset analysis

scRNA-seq data of patients with treatment-naïve lung adenocarcinoma were obtained from GSE131907 (30). Data processing, analysis, and visualization were conducted using R program with package Seurat. Only CD8+ T cells (original labels: CD8 low T, cytotoxic CD8+ T, naïve CD8+ T, and exhausted CD8+ T) with tissues from 11 tumor-containing lung tissues and 10 nonmetastatic lung-draining lymph nodes (original labels: tLung and nLN, respectively) were extracted and used for the analysis. CD8+ T cells were grouped into four clusters on the basis of the marker gene expressions: stemlike (TCF7+GZMBPDCD1+); terminally differentiated (TCF7GZMB+PDCD1+); naïve (TCF7+GZMBPDCD1); effector (TCF7GZMB+PDCD1) and visualized with UMAP.

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. The scRNA-seq and RNA-seq data reported in this paper have been deposited in Gene Expression Omnibus (GEO) under accession number GSE228386, GSE228387, and GSE228388.

Statistical analyses

Data were analyzed using the Graphpad Prism v.8.4.3 (GraphPad Software). The results of statistical analyses are indicated as the mean ± SD, unless otherwise indicated. Intergroup variance was assessed using the F-test. For normally distributed data, statistical significance was determined using an unpaired two-sided Student t test. An unpaired two-sided Welch t test was used to analyze groups that showed differing variance. For multiple comparisons, statistical significance was determined using a one-way ANOVA, followed by Dunnett test or Tukey test. P values of < 0.05 were considered statistically significant. Gene clustering was performed on the basis of Pearson correlation coefficient.

CD69 deficiency increases cytotoxic CD8+ T cells in the TME

To investigate the mechanism of enhanced antitumor immunity in Cd69KO mice, wherein tumor growth was significantly diminished (ref. 17; Fig. 1A), we utilized CT26 colorectal carcinoma cells that expressed AH1 antigen so that tumor-specific CD8+ T cells could be detected using an H-2Ld-AH1 tetramer (Supplementary Fig. S1A; ref. 31). We first analyzed tumor-infiltrating lymphocytes 14 days after tumor inoculation and found that CD69 deficiency increased the frequency of total CD8+ T cells (Fig. 1B; Supplementary Fig. S1B). Moreover, the frequency of AH1 tetramer+ tumor-specific CD8+ T cells was increased in Cd69KO mice (Fig. 1C). Almost all tumor-infiltrating AH1 tetramer+ CD8+ T cells in both Cd69WT and Cd69KO mice expressed PD-1 (Fig. 1D), whereas the frequency of granzyme B-expressing AH1 tetramer+ CD8+ T cells was significantly increased in Cd69KO mice (Fig. 1E). These data demonstrate that CD69 deficiency increases tumor-specific cytotoxic CD8+ T cells in the TME.

Figure 1.

CD69 controls the differentiation of intratumoral stemlike CD8+ T cells into functional terminally differentiated CD8+ T cells. A, Tumor growth curves. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. n = 5 per group; mean volume ± SEM. B, A schematic outline of CT26 tumor model. C–E, Intratumoral CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. At least n = 6 per group. Number indicates the percentage of gated cells. The frequency of AH1 tetramer+ CD8+ T cells in CD45+ cells are shown in representative flow plots (left) and a summary of the experiments (right; C). The frequency of PD-1+ cells in AH1 tetramer+ cells (D). The frequency of granzyme B+ cells in AH1 tetramer+PD-1+ cells (E). F and G, scRNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. Experiments were performed in duplicate. Pool of four mice in one group. A UMAP projection of Cd69WT and Cd69KO cells that were assigned to four clusters and color-coded on the basis of clusters, determined by Seurat (F). UMAP plots of Cd69WT and Cd69KO cells (left). The graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters (G). H and I, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after inoculation. n = 11 per group. The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells (H). The frequency of CD8+ T-cell subpopulations in CD45+ cells (I). J and K, Bulk RNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T-cell subpopulations were sorted at 14 days after CT26 cell inoculation and subjected to RNA-seq. Experiments were performed in duplicate. Pool of four mice in one group. The PCA plots of RNA-seq data of CD8+ T-cell subpopulations in Cd69WT and Cd69KO mice (J). A heat map showing the similarity and clustering of CD8+ T-cell subpopulations in Cd69WT and Cd69KO mice using gene expression profiles (K). A GSEA of the transcriptional signature from stemlike (left) or terminally differentiated (right) CD8+ T cells in B16-OVA melanoma in the ranked list of genes that were differentially expressed in AH1 tetramer+ stemlike CD8+ T cells in Cd69KO mice versus Cd69WT mice (L). A GSEA of the transcriptional signature from stemlike or terminally differentiated CD8+ T cells in LCMV clone-13 infection (M). Term diff, terminally differentiated. NES, normalized enrichment score. An unpaired two-sided Student t test or Welch t test was used for the analyses of flow cytometry and tumor growth curves. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. ns: not significant. Representative data from three experiments with at least five mice are shown (A, D and E). Data are pooled from two experiments with at least five mice (C, H and I).

Figure 1.

CD69 controls the differentiation of intratumoral stemlike CD8+ T cells into functional terminally differentiated CD8+ T cells. A, Tumor growth curves. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. n = 5 per group; mean volume ± SEM. B, A schematic outline of CT26 tumor model. C–E, Intratumoral CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. At least n = 6 per group. Number indicates the percentage of gated cells. The frequency of AH1 tetramer+ CD8+ T cells in CD45+ cells are shown in representative flow plots (left) and a summary of the experiments (right; C). The frequency of PD-1+ cells in AH1 tetramer+ cells (D). The frequency of granzyme B+ cells in AH1 tetramer+PD-1+ cells (E). F and G, scRNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. Experiments were performed in duplicate. Pool of four mice in one group. A UMAP projection of Cd69WT and Cd69KO cells that were assigned to four clusters and color-coded on the basis of clusters, determined by Seurat (F). UMAP plots of Cd69WT and Cd69KO cells (left). The graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters (G). H and I, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after inoculation. n = 11 per group. The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells (H). The frequency of CD8+ T-cell subpopulations in CD45+ cells (I). J and K, Bulk RNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T-cell subpopulations were sorted at 14 days after CT26 cell inoculation and subjected to RNA-seq. Experiments were performed in duplicate. Pool of four mice in one group. The PCA plots of RNA-seq data of CD8+ T-cell subpopulations in Cd69WT and Cd69KO mice (J). A heat map showing the similarity and clustering of CD8+ T-cell subpopulations in Cd69WT and Cd69KO mice using gene expression profiles (K). A GSEA of the transcriptional signature from stemlike (left) or terminally differentiated (right) CD8+ T cells in B16-OVA melanoma in the ranked list of genes that were differentially expressed in AH1 tetramer+ stemlike CD8+ T cells in Cd69KO mice versus Cd69WT mice (L). A GSEA of the transcriptional signature from stemlike or terminally differentiated CD8+ T cells in LCMV clone-13 infection (M). Term diff, terminally differentiated. NES, normalized enrichment score. An unpaired two-sided Student t test or Welch t test was used for the analyses of flow cytometry and tumor growth curves. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. ns: not significant. Representative data from three experiments with at least five mice are shown (A, D and E). Data are pooled from two experiments with at least five mice (C, H and I).

Close modal

CD69 controls the differentiation of intratumoral CD8+ T cells

To understand the impact of CD69 deficiency on intratumoral CD8+ T cells at a single-cell level, we performed an unbiased analysis of intratumoral tumor-specific CD8+ T cells using scRNA-seq and analyzed pooled scRNA-seq data of AH1-specific CD8+ T cells from Cd69WT and Cd69KO mice. UMAP and clustering analysis assigned cells into four clusters based on their transcriptomes (Fig. 1F), and the top 10 differentially expressed genes across the clusters were examined (Supplementary Fig. S2A–S2C). Cells in cluster 0 highly expressed Gzmb and Havcr2 (encoding Tim-3), representing the terminally differentiated CD8+ T-cell population. Cells in cluster 1 expressed high Tcf7 (encoding Tcf1) and Slamf6 (encoding Ly108), which represent the stemlike CD8+ T-cell population. Cells in clusters 2 and 3 exhibited the expression of proliferation markers, such as Birc5 (for G2–M phase) and Mcm6 (for S phase), which were confirmed by cell-cycle scoring (Supplementary Fig. S2D; ref. 23). Cd69WT and Cd69KO cells occupied distinct spaces in the UMAP plots; more terminally differentiated CD8+ T cells and fewer stemlike CD8+ T cells were detected in Cd69KO mice than in Cd69WT mice (Fig. 1G). Flow cytometry confirmed that the frequency of AH1-specific, Tim-3+ terminally differentiated CD8+ T cells was increased in Cd69KO mice, whereas Tcf1+ stemlike CD8+ T cells were decreased (Fig. 1H). To estimate the absolute cell number, we calculated the frequency of each cell population among CD45+ cells; Tim-3+ terminally differentiated CD8+ T cells were significantly increased in Cd69KO mice, whereas Tcf1+ stemlike CD8+ T cells did not change (Fig. 1I). In addition, we found that CD69 was expressed on both AH1-specific stemlike and terminally differentiated CD8+ T cells (Supplementary Fig. S2E). These data indicate that the number of tumor-specific terminally differentiated CD8+ T cells is increased in Cd69KO mice, whereas the proportion of stemlike CD8+ T cells is comparable between Cd69WT and Cd69KO mice.

CD69 suppresses the transcriptional program for terminal differentiation of CD8+ T cells

To investigate the mechanism behind the impact of CD69 deficiency on tumor-specific CD8+ T cells, we next assessed the gene expression profiles of individual AH1-specific, Ly108+ stemlike and Tim-3+ terminally differentiated CD8+ T-cells isolated from Cd69WT and Cd69KO mice (Supplementary Fig. S3A and S3B). Data were first compared with the datasets of antigen-specific CD8+ T-cell subpopulations (i.e., stemlike and terminally differentiated CD8+ T cells) obtained from B16-OVA melanoma-bearing mice and LCMV clone-13-infected mice, which were previously reported (1). A gene set enrichment analysis (GSEA) demonstrated significant transcriptional similarity between the corresponding subpopulations in both Cd69WT and Cd69KO mice (Supplementary Fig. S3C and S3D; Supplementary Table S1). However, the Cd69WT and Cd69KO CD8+ T-cell subpopulations exhibited a distinct transcriptome profile (Fig. 1J and K). Furthermore, Cd69KO Ly108+ stemlike CD8+ T-cells showed a gene expression profile with more signatures of terminally differentiated CD8+ T cells than Cd69WT Ly108+ stemlike CD8+ T cells (Fig. 1L and M). These data raise the possibility that CD69 suppresses the transcriptional program toward terminal differentiation in stemlike CD8+ T cells.

Low TOX expression in CD69-deficient CD8+ T cells in the TME

To test whether CD69 regulated the transcriptional program for differentiation of terminally differentiated CD8+ T cells, gene expression profiles of individual Ly108+ stemlike and Tim-3+ terminally differentiated CD8+ T cells were analyzed, and several differentially expressed genes between Cd69WT and Cd69KO mice were identified (Fig. 2A and B; Supplementary Fig. S4A and S4B; Supplementary Tables S2–S3). Three genes encoding transcription factors (i.e., Tox, Crem, and Irf8) were downregulated, whereas one gene (i.e., Satb1) was upregulated in both stemlike and terminally differentiated Cd69KO CD8+ T cells (Supplementary Fig. S4A and S4B; Supplementary Tables S2–S3). Among them, we focused on Tox because TOX has been reported to be important for the CD8+ T-cell function under conditions associated with chronic antigen stimulation, such as cancer or chronic viral infection (29, 32–34), and to be crucial for maintaining Tcf1+ stemlike CD8+ T cells (35). qRT-PCR (Supplementary Fig. S4C) and flow cytometry (Fig. 2C) confirmed low expression of TOX mRNA and protein in both Ly108+ stemlike and Tim-3+ terminally differentiated CD8+ T cells in the TME of Cd69KO mice. We also found that both stemlike and terminally differentiated CD8+ T cells in Cd69KO mice were significantly enriched in gene signatures from the corresponding subpopulations in TOX-deficient mice infected with LCMV clone-13 (Supplementary Fig. S4D and S4E; ref. 29). Furthermore, expression of inhibitory receptors, such as PD-1, Lag-3, and TIGIT, known as downstream molecules of TOX (29, 32, 33, 34), was significantly diminished in intratumoral CD8+ T-cell subpopulations in Cd69KO mice (Fig. 2D). These data demonstrate that the TOX-mediated transcriptional program is repressed in intratumoral CD8+ T cells in Cd69KO mice.

Figure 2.

CD69 controls the expression of the transcription factor TOX in tumor-specific CD8+ T cells. A, A schematic outline of CT26 tumor model. B, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T-cell subpopulations were sorted at 14 days after CT26 cell inoculation and subjected to RNA-seq. The MA plot depicts the differential gene expression in Ly108+ stemlike and Tim-3+ terminally differentiated CD8+ T cells in Cd69KO mice compared to Cd69WT mice. Log2 average expression > 2 is analyzed. Log2 fold change > 0.4 and ←0.4 is shown in red and blue, respectively. The average of duplicate samples were analyzed. Pool of four mice in one group. C and D, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. n = 6 per group. TOX MFI in CD8+ T-cell subpopulations is shown in a representative histogram (left) and a summary of the experiments (right; C). MFI of the indicated proteins in CD8+ T-cell subpopulations (D). Splenic CD8+ cells are shown as a control. E, A schematic outline of CT26 tumor model. F and G, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells in TDLNs and nonTDLN were analyzed at 14 days after inoculation. n = 6 per group. The frequency of AH1 tetramer+ cells in lymph node cells is shown (left) and the graph represents the absolute number of AH1 tetramer+ CD8+ T cells (F). MFI of the indicated proteins in AH1 tetramer+ CD8+ T cells (G). An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. ns: not significant. Representative data from three experiments with at least five mice are shown (C and D, F and G).

Figure 2.

CD69 controls the expression of the transcription factor TOX in tumor-specific CD8+ T cells. A, A schematic outline of CT26 tumor model. B, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T-cell subpopulations were sorted at 14 days after CT26 cell inoculation and subjected to RNA-seq. The MA plot depicts the differential gene expression in Ly108+ stemlike and Tim-3+ terminally differentiated CD8+ T cells in Cd69KO mice compared to Cd69WT mice. Log2 average expression > 2 is analyzed. Log2 fold change > 0.4 and ←0.4 is shown in red and blue, respectively. The average of duplicate samples were analyzed. Pool of four mice in one group. C and D, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. Intratumoral AH1 tetramer+ CD8+ T cells were analyzed at 14 days after CT26 cell inoculation. n = 6 per group. TOX MFI in CD8+ T-cell subpopulations is shown in a representative histogram (left) and a summary of the experiments (right; C). MFI of the indicated proteins in CD8+ T-cell subpopulations (D). Splenic CD8+ cells are shown as a control. E, A schematic outline of CT26 tumor model. F and G, CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells in TDLNs and nonTDLN were analyzed at 14 days after inoculation. n = 6 per group. The frequency of AH1 tetramer+ cells in lymph node cells is shown (left) and the graph represents the absolute number of AH1 tetramer+ CD8+ T cells (F). MFI of the indicated proteins in AH1 tetramer+ CD8+ T cells (G). An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. ns: not significant. Representative data from three experiments with at least five mice are shown (C and D, F and G).

Close modal

CD69 deficiency represses the induction of TOX in tumor-specific CD8+ T cells in TDLNs

It has been reported that TDLNs play a pivotal role in initiating antitumor CD8+ T-cell responses (6–9) and that the expression of TOX can be induced by T-cell receptor (TCR) stimulation (29, 32, 33). These facts prompted us to investigate the TOX levels in tumor-specific CD8+ T cells in TDLNs (Fig. 2E; Supplementary Fig. S5A). We first found that the number of AH1-specific CD8+ T cells in TDLNs from Cd69KO mice was significantly higher compared with Cd69WT mice, whereas AH1-specific CD8+ T cells were nearly absent in the non-TDLNs in both Cd69WT and Cd69KO mice (Fig. 2F). Expression of TOX and PD-1 in AH1 tetramer+ tumor-specific CD8+ T cells in TDLNs was significantly decreased in Cd69KO mice (Fig. 2G), whereas the expression of Lag-3 and TIGIT were very low on CD8+ T cells, even in Cd69WT mice, and there were no significant differences between Cd69WT and Cd69KO mice (Supplementary Fig. S5B). In addition, we confirmed that CD69 was expressed on AH1-specific CD8+ T cells in TDLNs (Supplementary Fig. S5C). These results demonstrate that CD69 deficiency diminishes the induction of TOX in TDLNs.

CD69 controls differentiation of CD8+ T cells in TDLNs

Because TOX expression in Cd69KO CD8+ T cells was already diminished in TDLNs led us to examine the characteristics of tumor-specific CD8+ T cells in TDLNs. We performed an unbiased analysis of AH1-specific CD8+ T cells in TDLNs by conducting scRNA-seq and analyzed pooled scRNA-seq data from Cd69WT and Cd69KO mice (Fig. 3A). Five clusters were identified on the basis of their transcriptomes (Fig. 3B), and the top 10 differentially expressed genes across the clusters were examined (Supplementary Fig. S6A and S6B). Cells in both clusters 0 and 1 expressed high Tcf7 and Slamf6, markers for the stemlike CD8+ T-cell population, indicating that there were two types of stemlike CD8+ T-cell subsets. Cells in cluster 2 expressed Gzmb and Havcr2, representing the terminally differentiated CD8+ T-cell population. Cells in cluster 3 represented the proliferating cell population because they exhibited the expression of proliferation markers (e.g., Birc5 and Mcm6), which is consistent with the cell-cycle scoring (Supplementary Fig. S6C). Cells in cluster 4 expressed Cd69, Ccr9, and Sell (encoding CD62L), representing that the early-activated T-cell population still maintained some naïve T-cell signatures. Importantly, the proportion of Cd69KO cells in each cluster was altered compared to Cd69WT cells; more terminally differentiated CD8+ T cells and fewer stemlike CD8+ T cells were detected in Cd69KO mice than in Cd69WT mice (Fig. 3C). Flow cytometry confirmed that AH1-specific, Tim-3+ terminally differentiated CD8+ T cells were present and that the frequency of AH1-specific, Tim-3+ terminally differentiated CD8+ T cells was significantly increased in Cd69KO mice (Fig. 3D). A time-course analysis revealed that Tim-3+ terminally differentiated CD8+ T cells were barely detectable eight days after tumor inoculation, although TOX expression was already decreased in Cd69KO mice, suggesting that the enhanced generation of Tim-3+ terminally differentiated CD8+ T cells was preceded by TOX downregulation in TDLNs (Supplementary Fig. S6D–S6F). Furthermore, we found that the downregulation of TOX was observed in both Eomes+ stemlike 1 and CD94+ stemlike 2 CD8+ T cells, as well as in Tim-3+ terminally differentiated CD8+ T cells in Cd69KO mice (Fig. 3E; Supplementary Fig. S5A), suggesting that CD69 controls the induction of TOX in the early-phases of tumor-specific CD8+ T-cell differentiation in TDLNs.

Figure 3.

CD69 controls the differentiation of tumor-specific CD8+ T cells in TDLNs. A, A schematic outline of CT26 tumor model. B and C, scRNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 6-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells were isolated from TDLNs at 14 days after inoculation. Experiments were performed in duplicate. Pool of 10 mice in one group. An UMAP projection of Cd69WT and Cd69KO cells that were assigned to five clusters and color-coded on the basis of the clusters, determined by Seurat (B). UMAP plots of Cd69WT and Cd69KO cells (left) and the graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters (C). D and E, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells in TDLNs were analyzed at 14 days after CT26 cell inoculation. n = 6 per group. The number indicates the percentage of gated cells. Splenic CD8+ cells are shown as a control. The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells shown in representative flow plots (left) and a summary of the experiments (right; D). TOX MFI in Eomes+ stemlike 1, CD94+ stemlike 2 and terminally differentiated CD8+ T cells in TDLNs (E). F, A schematic outline of the mixed CD8+ T-cell transfer model during MC38-OVA development. G and H, An equal mixture of naïve OT-I CD8+ T-cells collected from Cd69WT CD45.2 and Cd69KO CD45.1 12-week-old female C57BL/6 mice were intravenously injected into CD45.1/CD45.2 12-week-old female C57BL/6 mice on day -1. MC38-OVA cells were subcutaneously inoculated on the next day (day 0) and OT-I CD8+ T cells in TDLNs were analyzed at the indicated time. At least n = 4 per group. The frequency of CD8+ T-cell subpopulations in PD-1+ OT-I CD8+ T-cells derived from each genotype (G). TOX MFI in PD-1+ OT-I CD8+ T cells (H). An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Representative data from three experiments with at least four mice are shown (D, G and H). Representative data from two experiments with at least five mice are shown (E).

Figure 3.

CD69 controls the differentiation of tumor-specific CD8+ T cells in TDLNs. A, A schematic outline of CT26 tumor model. B and C, scRNA-seq. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 6-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells were isolated from TDLNs at 14 days after inoculation. Experiments were performed in duplicate. Pool of 10 mice in one group. An UMAP projection of Cd69WT and Cd69KO cells that were assigned to five clusters and color-coded on the basis of the clusters, determined by Seurat (B). UMAP plots of Cd69WT and Cd69KO cells (left) and the graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters (C). D and E, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 7-week-old female BALB/c mice. AH1 tetramer+ CD8+ T cells in TDLNs were analyzed at 14 days after CT26 cell inoculation. n = 6 per group. The number indicates the percentage of gated cells. Splenic CD8+ cells are shown as a control. The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells shown in representative flow plots (left) and a summary of the experiments (right; D). TOX MFI in Eomes+ stemlike 1, CD94+ stemlike 2 and terminally differentiated CD8+ T cells in TDLNs (E). F, A schematic outline of the mixed CD8+ T-cell transfer model during MC38-OVA development. G and H, An equal mixture of naïve OT-I CD8+ T-cells collected from Cd69WT CD45.2 and Cd69KO CD45.1 12-week-old female C57BL/6 mice were intravenously injected into CD45.1/CD45.2 12-week-old female C57BL/6 mice on day -1. MC38-OVA cells were subcutaneously inoculated on the next day (day 0) and OT-I CD8+ T cells in TDLNs were analyzed at the indicated time. At least n = 4 per group. The frequency of CD8+ T-cell subpopulations in PD-1+ OT-I CD8+ T-cells derived from each genotype (G). TOX MFI in PD-1+ OT-I CD8+ T cells (H). An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Representative data from three experiments with at least four mice are shown (D, G and H). Representative data from two experiments with at least five mice are shown (E).

Close modal

We further confirmed this using MC38 colorectal carcinoma cells expressing ovalbumin (MC38-OVA) by detecting the tumor-specific CD8+ T cells using H-2Kb-OVA tetramer (Supplementary Fig. S7A and S7B). OVA-specific CD8+ T-cell number in TDLNs from Cd69KO mice was significantly higher than that in Cd69WT mice (Supplementary Fig. S7C). CD69 deficiency increased the frequency of Tim-3+ terminally differentiated CD8+ T cells and decreased the frequency of Tcf1+ stemlike CD8+ T cells within TDLNs (Supplementary Fig. S7D). Moreover, reduced TOX expression was observed in both CD8+ T-cell subpopulations in Cd69KO mice (Supplementary Fig. S7E).

To determine the cell-intrinsic role of CD69 in CD8+ T-cell differentiation, we inoculated MC38-OVA into wild-type (WT) mice that had been previously treated with equal numbers of Cd69WT and Cd69KO naïve CD8+ T cells from OT-I TCR transgenic mice expressing OVA-specific TCR. We then analyzed the infused CD8+ T cells in TDLNs at the indicated times (Fig. 3F). Even in the same TDLN, the frequency of Cd69KO Tcf1+ stemlike OT-I CD8+ T cells was significantly decreased eight days after tumor inoculation, whereas the frequency of Cd69KO Tim-3+ terminally differentiated OT-I CD8+ T cells was inversely increased compared to Cd69WT cells (Fig. 3G). OVA-specific CD8+ T cells were likely more rapidly activated than AH1-specific CD8+ T cells in TDLNs (Supplementary Fig. S6E). Furthermore, the TOX expression in Cd69KO PD-1+ OT-I CD8+ T cells was also significantly decreased 8 days after tumor inoculation (Fig. 3H). These data indicate that CD69 regulates the induction of TOX in a cell-intrinsic manner, thereby controlling tumor-specific CD8+ T-cell differentiation in TDLNs.

We next investigated whether CD8+ T cells in TDLNs from patients with cancer expressed CD69 and TOX. To this end, we utilized a published scRNA-seq dataset from a study that included tumor-containing lung tissue (tLung) and nonmetastatic lung-draining lymph nodes (nLNs) collected from treatment-naïve patients with lung adenocarcinoma (30). CD8+ T cells from both tissues were grouped into four clusters (terminally differentiated, stemlike, effector, and naïve) and visualized by UMAP (Supplementary Fig. S8A and S8B). Importantly, both stemlike and terminally differentiated CD8+ T cells were present in nLNs of patients with cancer and expressed substantial levels of CD69 and TOX (Supplementary Fig. S8C and S8D).

Because dendritic cells (DC) play a central role in the priming of tumor-specific CD8+ T cells and because CD69 is known to control antigen presentation from monocytes (36, 37), we next tested whether CD69 played a role in the major DC populations in the TME and TDLNs (Supplementary Fig. S9A and S9B). We first observed that CD69 was expressed in all major DC populations in the TME and that the highest expression of CD69 was detected in monocyte-derived DCs (mo-DC; Supplementary Fig. S9C). In contrast, relatively low CD69 expression was observed in conventional DC1 (cDC1) and conventional DC2 (cDC2) in TDLNs (Supplementary Fig. S9C), and mo-DCs were not detected in TDLNs (Supplementary Fig. S9E and S9G).

We next evaluated whether CD69 deficiency influenced DCs (Supplementary Fig. S9D and S9E). The frequency of mo-DCs in CD45+ cells in the TME was increased in Cd69KO mice, suggesting that CD69 may control the number of mo-DCs (Supplementary Fig. S9D). Because the frequency of mo-DCs in TDLNs was very low in both Cd69WT and Cd69KO mice (Supplementary Fig. S9E), this increase in mo-DCs in Cd69KO mice may have occurred within the TME. We observed that the number of cDC1 and cDC2 was not changed between Cd69WT and Cd69KO mice in both the TME and TDLNs (Supplementary Fig. S9D and S9E), and no significant differences in the expression of costimulatory molecules, such as CD80 and CD86, were observed between Cd69WT and Cd69KO mice in DCs in the TME or TDLNs (Supplementary Fig. S9D and S9E). We next evaluated the effects of anti-CD69 administration on DCs in the TME and TDLNs. No significant differences in the frequency or phenotype of DCs were observed between anti-CD69–treated mice and control IgG-treated mice (Supplementary Fig. S9F and S9G).

These results suggest that CD69 deficiency and anti-CD69 treatment do not impact cDC1 and cDC2 in the TME and TDLNs. Because cDC1 and cDC2 are known to be major DC subsets activating tumor-specific CD8+ T cells, we conclude that antitumor DC function in Cd69KO mice was nearly intact.

CD69 controls CD8+ T-cell differentiation independently of the egress of T cells from TDLNs

We next asker how CD69 deficiency resulted in the low expression of TOX in TDLNs. We hypothesized that CD69 deficiency induced the surface expression of S1P1 so that T-cells leave early from the TDLN before upregulating the high levels of TOX, thus resulting in the low expression of TOX and efficient differentiation to terminally differentiated CD8+ T cells in the TME. However, this was unlikely based on the data. First, the frequency of AH1 tetramer+ CD8+ T cells was high, rather than low, in CD69KO mice (Fig. 2F), which was contrary to our expectation. Second, the frequency of Tim-3+ terminally differentiated CD8+ T cells was already increased in TDLNs in Cd69KO mice (Fig. 3C and D). Nevertheless, we administered an S1P receptor agonist, FTY720, during tumor development to examine the impact of the abrogation of T-cell exit from lymphoid organs (ref. 11; Fig. 4A). We first confirmed that the administration of FTY720 resulted in an increase of AH1-specific CD8+ T cells in TDLNs in both Cd69WT and Cd69KO mice (Fig. 4B), indicating that exit from the TDLNs required S1P1, as previously reported (8, 9). More Cd69KO cells egressed from the TDLNs than Cd69WT cells, as evidenced by cell accumulation in TDLNs with FTY720 treatment, which was more dramatic in Cd69KO mice than in Cd69WT mice. Nevertheless, the expression of TOX was again low in both stemlike and terminally differentiated CD8+ T cells in Cd69KO mice, even after FTY720 treatment (Fig. 4C), and the frequency of Tim-3+ terminally differentiated CD8+ T cells in Cd69KO mice was significantly higher than that in Cd69WT mice (Fig. 4D). These data demonstrate that the low expression of TOX in Cd69KO CD8+ T cells is not due to early exit from the TDLNs, and rather it is because of the loss of cell-intrinsic regulation by CD69.

Figure 4.

CD69 controls the differentiation of tumor-specific CD8+ T-cells independently of T-cell egress. A, A schematic outline of administration of FTY720 during CT26 tumor development. B–D, CT26 tumor-bearing Cd69WT and Cd69KO 10-week-old female BALB/c mice were treated with FTY720 to block lymphocyte migration. AH1 tetramer+ CD8+ T cells in TDLNs were analyzed at 14 days after CT26 cell inoculation. At least n = 5 per group. Splenic CD8+ cells are shown as a control. The number indicates the percentage of gated cells. The frequency of AH1 tetramer+ cells in CD45+ cells shown in representative flow plot (left) and the graph represents absolute number of AH1 tetramer+ CD8+ T cells (B). TOX MFI in CD8+ T-cell subpopulations (C). The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells (D). E, A schematic outline of CT26 tumor model. F, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. PD-1+ CD8+ T cells were isolated from TDLNs at 14 days after inoculation. MFI of nuclear NFAT2. Experiments were performed in triplicate. Pool of four mice in 1 group. An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Data are pooled from two experiments with at least four mice (B). Representative data from three experiments with at least five mice are shown (C and D). Representative data from three experiments (F).

Figure 4.

CD69 controls the differentiation of tumor-specific CD8+ T-cells independently of T-cell egress. A, A schematic outline of administration of FTY720 during CT26 tumor development. B–D, CT26 tumor-bearing Cd69WT and Cd69KO 10-week-old female BALB/c mice were treated with FTY720 to block lymphocyte migration. AH1 tetramer+ CD8+ T cells in TDLNs were analyzed at 14 days after CT26 cell inoculation. At least n = 5 per group. Splenic CD8+ cells are shown as a control. The number indicates the percentage of gated cells. The frequency of AH1 tetramer+ cells in CD45+ cells shown in representative flow plot (left) and the graph represents absolute number of AH1 tetramer+ CD8+ T cells (B). TOX MFI in CD8+ T-cell subpopulations (C). The frequency of CD8+ T-cell subpopulations in AH1 tetramer+PD-1+ CD8+ T cells (D). E, A schematic outline of CT26 tumor model. F, Flow cytometry. CT26 cells were subcutaneously inoculated into Cd69WT and Cd69KO 8-week-old female BALB/c mice. PD-1+ CD8+ T cells were isolated from TDLNs at 14 days after inoculation. MFI of nuclear NFAT2. Experiments were performed in triplicate. Pool of four mice in 1 group. An unpaired two-sided Student t test or Welch t test was used for the analysis of the flow cytometry data. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Data are pooled from two experiments with at least four mice (B). Representative data from three experiments with at least five mice are shown (C and D). Representative data from three experiments (F).

Close modal

It has been reported that the induction of TOX in CD8+ T-cells requires calcineurin/NFAT signaling (29, 32–35). We observed that NFAT2 (Nfatc1), but not NFAT1 (Nfatc2), was substantially expressed in AH1-specific CD8+ T cells in TDLNs (Supplementary Fig. S10A), which is consistent with the notion that NFAT2 mainly controls TOX expression in antigen-specific CD8+ T cells (32). Furthermore, nuclear NFAT2 was found to be decreased in PD-1+ CD8+ T cells in TDLNs in Cd69KO mice (Fig. 4E and F). We also confirmed that treatment with Cyclosporin A, an inhibitor of the calcineurin/NFAT pathway, decreased the TOX expression in tumor-specific CD69+CD8+ T cells in TDLNs (Supplementary Fig. S10B), suggesting that in vivo calcineurin/NFAT signaling indeed required TOX expression. We next examined whether CD69 deficiency influenced calcineurin/NFAT signaling using single-sample gene set variation analysis (ssGSVA). Constitutive active NFAT1 lacking AP-1-binding sites (CA-RIT-NFAT1) reportedly promotes Tox expression in CD8+ T cells (26, 34). Therefore, we used the gene set related to CA-RIT-NFAT1 (26) and found that it was depleted in Cd69KO AH1-specific CD8+ T cells in TDLNs (Supplementary Fig. S10C).

These data suggest that CD69-deficient CD8+ T cells reduce NFAT-dependent transactivation and leads to diminished TOX expression. We conclude that CD69 deficiency attenuates the expression of TOX in tumor-specific CD8+ T cells in TDLNs, most likely through an NFAT-dependent pathway, and thereby promotes the initiation of CD8+ T-cell differentiation in TDLNs, leading to enhanced antitumor immune responses.

Combined use of anti-CD69 and anti–PD-1 has an efficient therapeutic effect

With the aim of applying anti-CD69 and anti–PD-1 in the clinical setting, we first examined the effect of anti-CD69 administration on tumor-specific CD8+ T cells (Supplementary Fig. S11A). We confirmed that anti-CD69 resulted in reduced TOX expression inAH1-specific CD8+ T cells in TDLNs (Supplementary Fig. S11B). Furthermore, the generation of Tim-3+ terminally differentiated CD8+ T cells was enhanced by anti-CD69 treatment (Supplementary Fig. S11C). These results suggest that anti-CD69 treatment and CD69 deficiency exert similar effects on tumor-specific CD8+ T cells. We therefore next examined the differentiation of intratumoral CD8+ T cells (Fig. 5A). We pooled scRNA-seq data from intratumoral AH1-specific CD8+ T cells in mice treated with the indicated antibodies (Fig. 5B; Supplementary Fig. S11D–S11F). Anti-CD69 treatment increased Tim-3+ terminally differentiated CD8+ T cells, whereas anti–PD-1 treatment mainly increased Tcf1+ stemlike CD8+ T cells, which is consistent with the notion that anti–PD-1 therapy preferentially expands the stemlike subpopulation (refs. 1, 2; Fig. 5B). The combined use of anti-CD69 and anti–PD-1 increased both stemlike and terminally differentiated subpopulations (Fig. 5B). As a result, combined treatment with anti-CD69 and anti–PD-1 had an efficient therapeutic effect, whereas anti-CD69 or anti–PD-1 administration alone showed significant retardation of tumor growth (Fig. 5C). We also confirmed that anti-CD69 treatment resulted in the decreased expression of Tox in AH1-specific CD8+ T cells in the TME (Supplementary Fig. S11G).

Figure 5.

Anti-CD69 therapy promotes the generation of terminally differentiated CD8+ T cells and enhances the efficacy of anti–PD-1 therapy. A, A schematic outline of the administration of antibodies during CT26 tumor development. CT26 cells were subcutaneously inoculated into 7-week-old female BALB/c mice. Antibodies were intraperitoneally administered to mice twice a week, three times. B, scRNA-seq. Intratumoral AH1 tetramer+ CD8+ T cells were isolated at 14 days after inoculation. Experiments were performed in duplicate. Pool of four mice in one group. A UMAP projection of control IgG, anti-CD69, anti–PD-1, anti-CD69, and anti–PD-1 treated cells that were assigned to four clusters and color-coded on the basis of the clusters, determined by Seurat (left). The graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters. C, Tumor growth curve. CT26 cells were subcutaneously inoculated into 7-week-old female BALB/c mice. The indicated antibodies were then intraperitoneally administered to mice twice a week, 4 times. n = 5 per group; mean volume ± SEM. D, Tumor growth curve. B16-OVA cells were subcutaneously inoculated into 7-week-old C57BL/6 mice. Mice were treated as described in C. At least n = 6 per group; mean volume ± SEM. E, A schematic outline of the administration of antibodies during B16-OVA tumor development. B16-OVA cells were subcutaneously inoculated into 7-week-old female C57BL/6 mice. Antibodies were intraperitoneally administered to mice twice a week, 3 times. F–I, Flow cytometry. The number indicates the percentage of gated cells. n = 4 per group. The frequency of OVA tetramer+ cells in CD45+ cells shown in representative flow plot (left) and the graph represents absolute number of OVA tetramer+ CD8+ T cells in TDLNs (F) and the TME (G). The frequency of CD8+ T-cell subpopulations in OVA tetramer+PD-1+ CD8+ T cells in the TME shown in a representative flow plot (top) and the graph represents the frequency and absolute cell number of CD8+ T-cell subpopulations (bottom; H). The frequency of Granzyme B+ cells in OVA tetramer+PD-1+ CD8+ T cells shown in a representative flow plot (left) and the graph represents the frequency and absolute cell number of Granzyme B+ cells (right; I). A one-way ANOVA followed by Dunnett test for the analyses of flow cytometry and tumor growth curves. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Representative data from three experiments with at least five mice are shown (C). Data are pooled from two experiments with five mice (D). Representative data from two experiments with at least four mice are shown (F–I).

Figure 5.

Anti-CD69 therapy promotes the generation of terminally differentiated CD8+ T cells and enhances the efficacy of anti–PD-1 therapy. A, A schematic outline of the administration of antibodies during CT26 tumor development. CT26 cells were subcutaneously inoculated into 7-week-old female BALB/c mice. Antibodies were intraperitoneally administered to mice twice a week, three times. B, scRNA-seq. Intratumoral AH1 tetramer+ CD8+ T cells were isolated at 14 days after inoculation. Experiments were performed in duplicate. Pool of four mice in one group. A UMAP projection of control IgG, anti-CD69, anti–PD-1, anti-CD69, and anti–PD-1 treated cells that were assigned to four clusters and color-coded on the basis of the clusters, determined by Seurat (left). The graph represents the percentages of cells from the indicated cluster in each sample (right). The number indicates the percentage of stemlike and terminally differentiated clusters. C, Tumor growth curve. CT26 cells were subcutaneously inoculated into 7-week-old female BALB/c mice. The indicated antibodies were then intraperitoneally administered to mice twice a week, 4 times. n = 5 per group; mean volume ± SEM. D, Tumor growth curve. B16-OVA cells were subcutaneously inoculated into 7-week-old C57BL/6 mice. Mice were treated as described in C. At least n = 6 per group; mean volume ± SEM. E, A schematic outline of the administration of antibodies during B16-OVA tumor development. B16-OVA cells were subcutaneously inoculated into 7-week-old female C57BL/6 mice. Antibodies were intraperitoneally administered to mice twice a week, 3 times. F–I, Flow cytometry. The number indicates the percentage of gated cells. n = 4 per group. The frequency of OVA tetramer+ cells in CD45+ cells shown in representative flow plot (left) and the graph represents absolute number of OVA tetramer+ CD8+ T cells in TDLNs (F) and the TME (G). The frequency of CD8+ T-cell subpopulations in OVA tetramer+PD-1+ CD8+ T cells in the TME shown in a representative flow plot (top) and the graph represents the frequency and absolute cell number of CD8+ T-cell subpopulations (bottom; H). The frequency of Granzyme B+ cells in OVA tetramer+PD-1+ CD8+ T cells shown in a representative flow plot (left) and the graph represents the frequency and absolute cell number of Granzyme B+ cells (right; I). A one-way ANOVA followed by Dunnett test for the analyses of flow cytometry and tumor growth curves. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant. Representative data from three experiments with at least five mice are shown (C). Data are pooled from two experiments with five mice (D). Representative data from two experiments with at least four mice are shown (F–I).

Close modal

We further examined the therapeutic effect of anti-CD69 administration on the immunorefractory B16 melanoma tumor model (38). To monitor tumor-specific CD8+ T cells, we utilized B16-OVA cells so that the tumor-specific CD8+ T cells could be detected by H-2Kb-OVA tetramer (Supplementary Fig. S11H). Treatment with either anti-CD69 or anti–PD-1 administration alone was not associated with retardation of tumor growth, whereas combined treatment with anti-CD69 and anti–PD-1 had an enhanced therapeutic effect on melanoma (Fig. 5D).

To investigate the mechanism of enhanced antitumor immunity by combination treatment, we analyzed tumor-specific CD8+ T cells in both TDLNs and the TME (Fig. 5E). A flow cytometry analysis showed that the combined use of anti-CD69 and anti–PD-1 led to an increase in the number of OVA-specific CD8+ T cells in TDLNs (Fig. 5F). Consistent with this result, the number of tumor-specific CD8+ T cells in the TME was significantly increased by combination treatment (Fig. 5G). The frequency and number of Tim-3+ terminally differentiated CD8+ T cells were increased in mice treated with anti-CD69 and anti–PD-1 (Fig. 5H). The number of Tcf1+ stemlike CD8+ T cells did not change significantly, although the frequency of Tcf1+ stemlike CD8+ T cells was decreased by combination treatment (Fig. 5H). The frequency and number of granzyme B-expressing cells were also significantly increased by combination treatment (Fig. 5I). Taken together, these data demonstrate that anti-CD69 therapy promotes the generation of terminally differentiated CD8+ T cells and enhances antitumor immune responses that synergize with anti–PD-1 therapy.

The present study significantly enhances our understanding of how tumor-specific CD8+ T-cells differentiate into functional, Tim-3+ terminally differentiated CD8+ T cells. Our single-cell transcriptomics defined CD69 as an important regulator for the differentiation of tumor-specific CD8+ T cells. CD69 deficiency resulted in decreased expression of the transcription factor TOX in tumor-specific CD8+ T cells not only in the TME but also in TDLNs. Importantly, CD69-deficient mice showed enhanced antitumor immunity, with a higher number of functional Tim-3+ terminally differentiated CD8+ T cells in the TME. Furthermore, anti-CD69 treatment enhanced the generation of Tim-3+ terminally differentiated CD8+ T cells with enhanced antitumor function and also synergized with anti–PD-1 treatment to render immunorefractory B16 melanoma susceptible to immunotherapy.

An important finding in this study is that CD69 controls TOX expression in tumor-specific CD8+ T cells. Our data showed that CD69-deficient tumor-specific CD8+ T cells had reduced levels of TOX and enhanced antitumor immunity. Interestingly, Khan, and colleagues shows that tumor-specific CD8+ T cells with heterozygous deletion in the Tox gene have enhanced antitumor effects compared with WT CD8+ T cells that recognize same antigen (32). This data suggests that partial downregulation of TOX has a beneficial effect on antitumor immunity, although complete deletion of the Tox gene leads to a defect in the persistence of antigen-specific CD8+ T cells (29, 32–35). Furthermore, TOX is known to be critical for the maintenance of Tcf1+ stemlike CD8+ T cells, therefore, reduced TOX expression may render Tcf1+ stemlike CD8+ T cells susceptible to differentiation. Indeed, we observed that the generation of Tim-3+ terminally differentiated CD8+ T cells was increased in Cd69KO mice, which may account for the augmentation of antitumor immunity. Although the molecular mechanism underlying CD69-mediated TOX induction remains unclear, one possibility is that CD69 contributes to signaling through the TCR because nuclear NFAT2 was found to be decreased in Cd69KO mice. Consistent with this result, our ssGSVA suggested that CD69-deficient CD8+ T cells had reduced NFAT-dependent transactivation in TDLNs. Nevertheless, further study is needed to elucidate the detailed molecular mechanisms.

CD69 has a large extracellular domain that is said to interact with ligand molecules (10). Indeed, Myl9/12, Galectin-1, S100A8/A9, and oxidized low-density lipoprotein have been identified as potential CD69 ligands (13, 39–41). However, their contribution to the CD69 function in antitumor immunity remains unknown. The fact that anti-CD69 exerts antitumor effect suggests the involvement of the ligand-receptor interaction. Because we observed the phenotype of CD69 deficiency in tumor-specific CD8+ T cells in TDLNs, we think that some CD69 ligands may be supplied in cells localized in TDLNs. Nevertheless, screening of known and unknown ligands that regulate the CD69-mediated induction of TOX should be performed in the future, which may reveal the detailed molecular mechanisms underlying the CD69 function.

Another important finding in this study is that Tim-3+ terminally differentiated CD8+ T cells could be generated within TDLNs and that CD69 regulated this differentiation process by controlling TOX expression, which contrasts with recent studies reporting that Tim-3+ terminally differentiated CD8+ T cells are generated from Tcf1+ stemlike CD8+ T cells within the TME (1, 2, 4, 5). Our data support two nonmutually exclusive models of the generation of intratumoral Tim-3+ terminally differentiated CD8+ T cells: Tim-3+ terminally differentiated CD8+ T cells generated from Tcf1+ stemlike CD8+ T cells in TDLNs directly migrate into the TME and Tcf1+ stemlike CD8+ T cells in TDLNs directly migrate into the TME and then efficiently give rise to Tim-3+ terminally differentiated CD8+ T cells. In both cases, the fact that CD69 deficiency attenuated the expression of TOX in tumor-specific CD8+ T cells in TDLNs suggests that CD69 imposed tumor-specific CD8+ T-cell fate in TDLNs by regulating TOX expression.

One of the most significant findings of this study is that anti-CD69 treatment increased the generation of Tim-3+ terminally differentiated CD8+ T cells and thereby enhanced antitumor immune responses. Because anti–PD-1 therapy preferentially increased the frequency of Tcf1+ stemlike CD8+ T cells (1, 2), the combination therapies of anti-CD69 and anti–PD-1 are compatible and showed efficient therapeutic effect even on immunorefractory B16 melanoma. Because CD69 is expressed on not only T cells but also almost every leukocyte, side effects from anti-CD69 administration may be unavoidable (10). However, mice lacking whole-body CD69 are healthy and show no severe defects. We therefore predict that anti-CD69–associated adverse events will be tolerable. In conclusion, we showed that CD69 plays a crucial role in antitumor immunity by controlling CD8+ T-cell differentiation. Because CD69 is frequently expressed on the surface of CD8+ T cells in TDLNs in human cancers (14), our study provides the rationale for clinical evaluation of humanized monoclonal antibody to human CD69 as a new and feasible strategy for cancer immunotherapy.

A. Onodera reports grants from Japan Society for the Promotion of Science (JPSP) during the conduct of the study; grants from JSPS and grants from Astellas Foundation for Research on Metabolic Disorders (AFRMD) outside the submitted work. No disclosures were reported by the other authors.

R. Koyama-Nasu: Conceptualization, data curation, supervision, funding acquisition, investigation, writing–review, and editing. M.Y. Kimura: Conceptualization, data curation, supervision, funding acquisition, investigation, writing–review, and editing. M. Kiuchi: Investigation. A. Aoki: Investigation. Y. Wang: Investigation. Y. Mita: Investigation. I. Hasegawa: Investigation. Y. Endo: Investigation. A. Onodera: Investigation. K. Hirahara: Investigation. S. Motohashi: Investigation. T. Nakayama: Supervision.

This work was supported by the following grants: Ministry of Education, Culture, Sports, Science, and Technology (MEXT Japan) Grants-in-Aid for Scientific Research (S) 19H05650, (B) 20H03464, 18H02892, (C) 21K07234, 20K07652, 18K07257, Challenging Research (Pioneering) 18H05375, (Exploratory) 20K21537, 18K19466, Innovative Areas 19H04800; Grants-in-Aid for Transformative Research Areas (A) JP22H05189; Project for Cancer Research and Therapeutic Evolution (P-CREATE) from the Japan Agency for Medical Research and development (AMED) JP18cm0106339, JP20cm01106372; AMED-PRIME JP21gm6310024; AMED JP223fa627003. We thank Drs. Alfred Singer and Damon Tumes for their critical reading of the manuscript; Dr. T. Nakatsura (National Cancer Center Japan) for the B16-OVA cells; the NIH Tetramer facility for the tetramer reagents; and K. Sugaya for their expert aid in flow cytometry.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/).

1.
Miller
BC
,
Sen
DR
,
Al Abosy
R
,
Bi
K
,
Virkud
YV
,
LaFleur
MW
, et al
.
Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade
.
Nat Immunol
2019
;
20
:
326
36
.
2.
Siddiqui
I
,
Schaeuble
K
,
Chennupati
V
,
Fuertes Marraco
SA
,
Calderon-Copete
S
,
Pais Ferreira
D
, et al
.
Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy
.
Immunity
2019
;
50
:
195
211
.
3.
Brummelman
J
,
Mazza
EMC
,
Alvisi
G
,
Colombo
FS
,
Grilli
A
,
Mikulak
J
, et al
.
High-dimensional single cell analysis identifies stem-like cytotoxic CD8(+) T cells infiltrating human tumors
.
J Exp Med
2018
;
215
:
2520
35
.
4.
LaFleur
MW
,
Nguyen
TH
,
Coxe
MA
,
Miller
BC
,
Yates
KB
,
Gillis
JE
, et al
.
PTPN2 regulates the generation of exhausted CD8(+) T cell subpopulations and restrains tumor immunity
.
Nat Immunol
2019
;
20
:
1335
47
.
5.
Jansen
CS
,
Prokhnevska
N
,
Master
VA
,
Sanda
MG
,
Carlisle
JW
,
Bilen
MA
, et al
.
An intra-tumoral niche maintains and differentiates stem-like CD8 T cells
.
Nature
2019
;
576
:
465
70
.
6.
Dammeijer
F
,
van Gulijk
M
,
Mulder
EE
,
Lukkes
M
,
Klaase
L
,
van den Bosch
T
, et al
.
The PD-1/PD-L1-checkpoint restrains t cell immunity in tumor-draining lymph nodes
.
Cancer Cell
2020
;
38
:
685
700
.
7.
Buchwald
ZS
,
Nasti
TH
,
Lee
J
,
Eberhardt
CS
,
Wieland
A
,
Im
SJ
, et al
.
Tumor-draining lymph node is important for a robust abscopal effect stimulated by radiotherapy
.
J Immunother Cancer
2020
;
8
:
e000867
.
8.
Connolly
KA
,
Kuchroo
M
,
Venkat
A
,
Khatun
A
,
Wang
J
,
William
I
, et al
.
A reservoir of stem-like CD8(+) T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response
.
Sci Immunol
2021
;
6
:
eabg7836
.
9.
Schenkel
JM
,
Herbst
RH
,
Canner
D
,
Li
A
,
Hillman
M
,
Shanahan
SL
, et al
.
Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1(+) CD8(+) T cells in tumor-draining lymph nodes
.
Immunity
2021
;
54
:
2338
53
.
10.
Kimura
MY
,
Koyama-Nasu
R
,
Yagi
R
,
Nakayama
T
.
A new therapeutic target: the CD69-Myl9 system in immune responses
.
Semin Immunopathol
2019
;
41
:
349
58
.
11.
Cyster
JG
,
Schwab
SR
.
Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs
.
Annu Rev Immunol
2012
;
30
:
69
94
.
12.
Ziegler
SF
,
Ramsdell
F
,
Hjerrild
KA
,
Armitage
RJ
,
Grabstein
KH
,
Hennen
KB
, et al
.
Molecular characterization of the early activation antigen CD69: a type II membrane glycoprotein related to a family of natural killer cell activation antigens
.
Eur J Immunol
1993
;
23
:
1643
8
.
13.
Hayashizaki
K
,
Kimura
MY
,
Tokoyoda
K
,
Hosokawa
H
,
Shinoda
K
,
Hirahara
K
, et al
.
Myosin light chains 9 and 12 are functional ligands for CD69 that regulate airway inflammation
.
Sci Immunol
2016
;
1
:
eaaf9154
.
14.
Piersiala
K
,
Farrajota Neves da Silva
P
,
Hjalmarsson
E
,
Kolev
A
,
Kagedal
A
,
Starkhammar
M
, et al
.
CD4(+) and CD8(+) T cells in sentinel nodes exhibit distinct pattern of PD-1, CD69, and HLA-DR expression compared to tumor tissue in oral squamous cell carcinoma
.
Cancer Sci
2021
;
112
:
1048
59
.
15.
Esplugues
E
,
Sancho
D
,
Vega-Ramos
J
,
Martinez
C
,
Syrbe
U
,
Hamann
A
, et al
.
Enhanced antitumor immunity in mice deficient in CD69
.
J Exp Med
2003
;
197
:
1093
106
.
16.
Esplugues
E
,
Vega-Ramos
J
,
Cartoixa
D
,
Vazquez
BN
,
Salaet
I
,
Engel
P
, et al
.
Induction of tumor NK-cell immunity by anti-CD69 antibody therapy
.
Blood
2005
;
105
:
4399
406
.
17.
Mita
Y
,
Kimura
MY
,
Hayashizaki
K
,
Koyama-Nasu
R
,
Ito
T
,
Motohashi
S
, et al
.
Crucial role of CD69 in anti-tumor immunity through regulating the exhaustion of tumor-infiltrating T cells
.
Int Immunol
2018
;
30
:
559
67
.
18.
Wei
SM
,
Pan
HL
,
Wang
L
,
Yin
GL
,
Zhong
K
,
Zhou
Y
, et al
.
Combination therapy with dendritic cell-based vaccine and anti-CD69 antibody enhances antitumor efficacy in renal cell carcinoma-bearing mice
.
Turk J Med Sci
2017
;
47
:
658
67
.
19.
Murata
K
,
Inami
M
,
Hasegawa
A
,
Kubo
S
,
Kimura
M
,
Yamashita
M
, et al
.
CD69-null mice protected from arthritis induced with anti-type II collagen antibodies
.
Int Immunol
2003
;
15
:
987
92
.
20.
Ito
T
,
Hirahara
K
,
Onodera
A
,
Koyama-Nasu
R
,
Yano
I
,
Nakayama
T
.
Anti-tumor immunity via the superoxide-eosinophil axis induced by a lipophilic component of Mycobacterium lipomannan
.
Int Immunol
2017
;
29
:
411
21
.
21.
Podtschaske
M
,
Benary
U
,
Zwinger
S
,
Hofer
T
,
Radbruch
A
,
Baumgrass
R
.
Digital NFATc2 activation per cell transforms graded T cell receptor activation into an all-or-none IL-2 expression
.
PLoS One
2007
;
2
:
e935
.
22.
Macosko
EZ
,
Basu
A
,
Satija
R
,
Nemesh
J
,
Shekhar
K
,
Goldman
M
, et al
.
Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets
.
Cell
2015
;
161
:
1202
14
.
23.
Tirosh
I
,
Izar
B
,
Prakadan
SM
,
Wadsworth
MH
, 2nd,
Treacy
D
,
Trombetta
JJ
, et al
.
Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq
.
Science
2016
;
352
:
189
96
.
24.
Alquicira-Hernandez
J
,
Powell
JE
.
Nebulosa recovers single cell gene expression signals by kernel density estimation
.
Bioinformatics
2021
;
37
:
2485
7
.
25.
Hanzelmann
S
,
Castelo
R
,
Guinney
J
.
GSVA: gene set variation analysis for microarray and RNA-seq data
.
BMC Bioinf
2013
;
14
:
7
.
26.
Martinez
GJ
,
Pereira
RM
,
Aijo
T
,
Kim
EY
,
Marangoni
F
,
Pipkin
ME
, et al
.
The transcription factor NFAT promotes exhaustion of activated CD8(+) T cells
.
Immunity
2015
;
42
:
265
78
.
27.
Onodera
A
,
Tumes
DJ
,
Watanabe
Y
,
Hirahara
K
,
Kaneda
A
,
Sugiyama
F
, et al
.
Spatial interplay between polycomb and trithorax complexes controls transcriptional activity in T lymphocytes
.
Mol Cell Biol
2015
;
35
:
3841
53
.
28.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
29.
Alfei
F
,
Kanev
K
,
Hofmann
M
,
Wu
M
,
Ghoneim
HE
,
Roelli
P
, et al
.
TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection
.
Nature
2019
;
571
:
265
9
.
30.
Kim
N
,
Kim
HK
,
Lee
K
,
Hong
Y
,
Cho
JH
,
Choi
JW
, et al
.
Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma
.
Nat Commun
2020
;
11
:
2285
.
31.
Castle
JC
,
Loewer
M
,
Boegel
S
,
de Graaf
J
,
Bender
C
,
Tadmor
AD
, et al
.
Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma
.
BMC Genomics
2014
;
15
:
190
.
32.
Khan
O
,
Giles
JR
,
McDonald
S
,
Manne
S
,
Ngiow
SF
,
Patel
KP
, et al
.
TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion
.
Nature
2019
;
571
:
211
8
.
33.
Scott
AC
,
Dundar
F
,
Zumbo
P
,
Chandran
SS
,
Klebanoff
CA
,
Shakiba
M
, et al
.
TOX is a critical regulator of tumour-specific T cell differentiation
.
Nature
2019
;
571
:
270
4
.
34.
Seo
H
,
Chen
J
,
Gonzalez-Avalos
E
,
Samaniego-Castruita
D
,
Das
A
,
Wang
YH
, et al
.
TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8(+) T cell exhaustion
.
Proc Natl Acad Sci U S A
2019
;
116
:
12410
5
.
35.
Yao
C
,
Sun
HW
,
Lacey
NE
,
Ji
Y
,
Moseman
EA
,
Shih
HY
, et al
.
Single-cell RNA-seq reveals TOX as a key regulator of CD8(+) T cell persistence in chronic infection
.
Nat Immunol
2019
;
20
:
890
901
.
36.
Fu
C
,
Jiang
A
.
Dendritic cells and CD8 T cell immunity in tumor microenvironment
.
Front Immunol
2018
;
9
:
3059
.
37.
Baeyens
A
,
Bracero
S
,
Chaluvadi
VS
,
Khodadadi-Jamayran
A
,
Cammer
M
,
Schwab
SR
.
Monocyte-derived S1P in the lymph node regulates immune responses
.
Nature
2021
;
592
:
290
5
.
38.
Aranda
F
,
Llopiz
D
,
Diaz-Valdes
N
,
Riezu-Boj
JI
,
Bezunartea
J
,
Ruiz
M
, et al
.
Adjuvant combination and antigen targeting as a strategy to induce polyfunctional and high-avidity T-cell responses against poorly immunogenic tumors
.
Cancer Res
2011
;
71
:
3214
24
.
39.
de la Fuente
H
,
Cruz-Adalia
A
,
Martinez Del Hoyo
G
,
Cibrian-Vera
D
,
Bonay
P
,
Perez-Hernandez
D
, et al
.
The leukocyte activation receptor CD69 controls T cell differentiation through its interaction with galectin-1
.
Mol Cell Biol
2014
;
34
:
2479
87
.
40.
Lin
CR
,
Wei
TY
,
Tsai
HY
,
Wu
YT
,
Wu
PY
,
C
ST
.
Glycosylation-dependent interaction between CD69 and S100A8/S100A9 complex is required for regulatory T-cell differentiation
.
FASEB J
2015
;
29
:
5006
17
.
41.
Tsilingiri
K
,
de la Fuente
H
,
Relano
M
,
Sanchez-Diaz
R
,
Rodriguez
C
,
Crespo
J
, et al
.
Oxidized low-density lipoprotein receptor in lymphocytes prevents atherosclerosis and predicts subclinical disease
.
Circulation
2019
;
139
:
243
55
.