Abstract Background T-cell exhaustion is a major barrier to effective antitumor immunity and limits the efficacy of cancer immunotherapies. This study investigates the role of suppressor of T-cell signaling 2 (STS2, also known as UBASH3A) in regulating CD8^+ T-cell exhaustion within the tumor microenvironment. Methods We used genetic ablation of STS2 in mouse models to assess tumor control and responses to anti-programmed cell death protein 1 (PD-1) checkpoint blockade therapy. CD8^+ tumor-infiltrating lymphocytes (TILs) were characterized through flow cytometry, mass cytometry, and single-cell transcriptomics. Mechanistic studies included co-immunoprecipitation, protein degradation assays, and endocytosis measurements to elucidate the interplay between STS2 and PD-1. Results STS2 expression progressively increased with T-cell exhaustion. Genetic deletion of STS2 enhanced tumor control and improved responses to anti-PD-1 therapy. STS2-deficient CD8^+ TILs maintained a more functional state, exhibiting enhanced effector activity, proliferation, and antitumor efficacy while resisting terminal exhaustion. Mechanistically, we discovered that STS2 physically interacts with PD-1 and modulates its expression, endocytosis, and degradation at the protein level. Conclusions Our findings establish STS2 as a multifaceted regulator of T-cell exhaustion and highlight its potential as a therapeutic target for enhancing antitumor immunity and improving cancer immunotherapy outcomes. Keywords: Immunosuppression, Immunotherapy __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * T-cell exhaustion is a major barrier to effective antitumor immunity and limits the efficacy of cancer immunotherapies. The molecular mechanisms underlying T-cell exhaustion in the tumor microenvironment are not fully understood. Suppressor of T-cell signaling 2 (STS2) is known to negatively regulate T-cell receptor signaling, but its role in T-cell exhaustion and antitumor immunity has not been explored. WHAT THIS STUDY ADDS * This study identifies STS2 as a critical regulator of CD8^+ T-cell differentiation in the tumor microenvironment. STS2 deficiency enhances tumor control and improves responses to anti-programmed cell death protein 1 (PD-1) checkpoint blockade therapy. STS2-deficient CD8^+ tumor-infiltrating lymphocytes maintain a more effective state, exhibiting enhanced functionality, proliferation, and antitumor activity while resisting terminal exhaustion. We uncover a novel mechanism by which STS2 regulates PD-1 expression at both transcriptional and post-transcriptional levels. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * These findings highlight STS2 as a potential therapeutic target for enhancing antitumor immunity and improving cancer immunotherapy outcomes. The study provides new insights into the molecular mechanisms of T-cell differentiation, which could inform the development of novel strategies to overcome resistance to checkpoint blockade therapies. Future research may focus on developing specific STS2 inhibitors and evaluating their efficacy in combination with existing immunotherapies. Introduction Cancer immunotherapy, particularly immune checkpoint blockade (ICB), has revolutionized cancer treatment in recent years, demonstrating unprecedented efficacy in various malignancies.[49]^1 CD8^+ T cells play a pivotal role in antitumor immunity, recognizing and eliminating cancer cells through their cytotoxic activity.[50]^2 However, in the tumor microenvironment (TME), these cells often become exhausted, limiting their ability to control tumor growth.[51]^3 Recent studies have revealed that T-cell exhaustion is a gradual process, with cells transitioning through various states from effector to terminally exhausted phenotypes.[52]^4 Importantly, cells in intermediate states of exhaustion, often referred to as “progenitor exhausted” T cells, retain some proliferative capacity and responsiveness to checkpoint blockade, making them a key target for therapeutic interventions.[53]^5 Understanding the molecular mechanisms underlying T-cell exhaustion is crucial for developing strategies to enhance the efficacy of cancer immunotherapies. Suppressor of T-cell receptor signaling 2 (STS2), also known as Ubiquitin-Associated And SH3 Domain-Containing Protein A (UBASH3A) and T-Cell Ubiquitin Ligand Protein,[54]^6 is a member of the STS family of proteins known to negatively regulate T-cell receptor (TCR) signaling.[55]^7 STS2 has been implicated in various autoimmune diseases[56]^8 9 and is highly expressed in lymphocytes.[57]^10 11 While its role in modulating TCR signaling has been established, the potential involvement of STS2 in T-cell exhaustion and antitumor immunity remains unexplored. Programmed cell death protein 1 (PD-1) is a key inhibitory receptor expressed on exhausted T cells and a major target for checkpoint blockade therapies.[58]^12 The regulation of PD-1 involves complex mechanisms at both transcriptional and post-translational levels.[59]^13 Recent studies have highlighted the importance of protein trafficking, endocytosis, and degradation in modulating PD-1 function.[60]^14 15 However, the potential role of STS2 in regulating PD-1 expression and function has not been investigated. In this study, we sought to elucidate the role of STS2 in CD8^+ T-cell exhaustion and antitumor immunity. We hypothesized that STS2, given its known inhibitory effects on T-cell signaling, might play a crucial role in promoting T-cell exhaustion in the TME. Using a combination of genetic mouse models, transcriptomic analyses, and functional assays, we investigated the impact of STS2 deficiency on CD8^+ T-cell function, exhaustion states, and antitumor responses. Furthermore, we explored the molecular mechanisms by which STS2 regulates T-cell function, with a particular focus on its interaction with PD-1 and its effects on protein metabolic process. Results STS2 is upregulated in tumor-infiltrating CD8^+ Tex cells While STS2 (gene symbol UBASH3A) is known to be highly expressed in peripheral blood and splenic lymphocytes, particularly T cells, under normal physiological conditions, its role in tumor development and its expression pattern within the TME remain unclear. To elucidate the immune cell expression of STS2 within the TME, we analyzed publicly available single-cell RNA-sequencing (scRNA-seq) data from human breast cancer tissue.[61]^16 Our analysis revealed high STS2 expression in T cells ([62]figure 1A). To validate these findings at the protein level, we performed CyTOF (Cytometry by time of flight) analysis of CD45-positive tumor-infiltrating cells in endometrial cancer tissues and found that STS2 is predominantly expressed in CD8^+ T cells, with detectable expression also observed in CD4^+ T cells ([63]online supplemental figure S1C,D). For further validation, we employed multiplex immunofluorescence on tissue microarray (TMA) of endometrial and colorectal cancers labeled with panCK/CD4/CD8/STS2 ([64]online supplemental figure S1E). In the endometrial cancer samples, STS2 expression was predominantly observed in CD8^+ T cells compared with CD4^+ T cells ([65]figure 1B). Similarly, in colorectal cancer TMAs, the proportion of STS2^+CD8^+ T cells was significantly higher than that of STS2^+CD4^+ T cells in both tumor and adjacent tissues ([66]figure 1C, [67]online supplemental figure S1F). Notably, the proportion of STS2^+CD8^+ T cells among all CD8^+ T cells was significantly higher in tumors compared with adjacent tissues, while STS2^+ cells among CD4^+ T cells showed only an upward trend in tumors ([68]online supplemental figure S1G). Figure 1. STS2 is upregulated in tumor-infiltrating CD8^+ Tex cells. (A) STS2 (gene symbol UBASH3A) mRNA levels in human breast cancer cohorts. (B–C) Immunofluorescent staining of indicated molecules and quantification of STS2^+CD4^+ or CD8^+ T cells of all CD4^+ or CD8^+ T cells in endometrial cancer (B, n=189) and colon cancer (C, n=156) tissues. The scale bar of higher magnification is 5 µm and the lower is 50 µm, as indicated in the images. Statistical analysis was done by paired Student’s t-test. (D) STS2 mRNA levels in CD8^+TILs as classified by ProjecTILs, across human (left) and mice (right) cohorts. (E) TCGA database RNA sequencing analysis of the Pearson correlation between the expression of STS2 and exhausted T-cell or effector T-cell signature genes in people with colon cancer (COAD), endometrial cancer (UCEC), breast cancer (BRCA), or skin cutaneous melanoma (SKCM). (F) t-SNE plots of CyTOF data depicting STS2 distribution across PhenoGraph-defined CD8^+ TIL populations in human endometrial cancer specimens. (G) STS2 expression per mouse (right) and representative histograms (left) in Tim3^−CD39^−, Tim3^+CD39^+ and all CD8^+ TILs (n=4 mice). Statistical analysis was done by paired Student’s t-test. (H-I) Effect of IL-2 on STS2 mRNA and protein expression after stimulation for 3 days in mice CD8^+T cells. Statistical analysis was done by Student’s t-test. (J) ATAC-seq and Cut&Run (H3K27ac)[69]^23 tracks of Stat5CA overexpression P14 cells at the STS2 locus. Blue highlights indicate ATAC peaks increased in P14 STAT5CA cells compared with P14 Empty. CM, central memory; CyTOF, cytometry by time of flight; EM, effector memory; IL, interleukin; MFI, mean fluorescent intensity; mRNA, messenger RNA; STS2, suppressor of T-cell receptor signaling 2 ;TCGA, The Cancer Genome Atlas; TEMRA, effector memory-expressing CD45RA T cells; Tex, exhausted T cells; TIL, tumor-infiltrating lymphocyte; Tpex, progenitor exhausted T cells; TPM, transcripts per million; t-SNE, t-distributed stochastic neighbor embedding. [70]Figure 1 [71]Open in a new tab To further characterize STS2 expression in various subtypes of tumor-infiltrating CD8^+ T cells, we analyzed publicly available scRNA-seq data from human and mouse tumor-infiltrating lymphocytes (TILs) atlas using ProjecTILs classification.[72]17,[73]19 We observed that STS2 expression gradually increased with CD8^+ T-cell differentiation state, with the highest expression in exhausted T cells (Tex), followed by progenitor exhausted T cells (Tpex) ([74]figure 1D). Transcriptomic data from TCGA (The Cancer Genome Atlas) cohorts (colon cancer (COAD), endometrial cancer (UCEC), breast cancer (BRCA), and skin cutaneous melanoma (SKCM)) showed significant positive correlations between STS2 expression and exhausted T-cell signatures (HAVCR2, TIGIT, LAG3, PDCD1, LAYN), stronger than those with effector T-cell signatures (CX3CR1, FGFBP2, FCGR3A) ([75]figure 1E). To validate STS2 expression in CD8^+ TILs, we performed CyTOF analysis on CD8^+ TILs from human endometrial cancer tissues and mouse MC38 tumors. In human endometrial cancer, STS2 expression was particularly enriched in exhausted CD8^+ T cells ([76]figure 1F, [77]online supplemental figure S2A). Analysis of CD8^+ TILs from murine MC38 tumors confirmed higher STS2 expression in terminally exhausted CD8^+ T cells (Tim3^+CD39^+) compared with Tim3^−CD39^− cells ([78]figure 1G). In transcriptional profiles from patients with melanoma treated with pembrolizumab,[79]^20 STS2 expression in tumors was higher than in PBMCs in non-naïve CD8^+ T cells ([80]online supplemental figure S2B). Analysis of tumor-infiltrating CD8^+ T cells transcriptome data from [81]GSE123235[82]^5 revealed significantly higher STS2 expression in terminally exhausted (Slamf6^−Tim3^+) cells compared with progenitor exhausted (Slamf6^+Tim3^−) cells ([83]online supplemental figure S2C, left). Similarly, in [84]GSE149876 data comparing four CD8^+ T-cell exhaustion states,[85]^21 STS2 expression was significantly higher in the terminal exhaustion group compared with progenitor and intermediate exhaustion groups ([86]online supplemental figure S2C, right). We further validated the high expression of STS2 in Tex cells using TISCH analysis of 14 public pan-cancer single-cell transcriptome datasets ([87]online supplemental figure S2D). To investigate the mechanism underlying STS2 upregulation, we found that interleukin (IL)-2 promoted STS2 expression in a dose-dependent manner at messenger RNA (mRNA) and protein levels ([88]figure 1H,I). Furthermore, STAT5 is a key downstream effector molecule of IL-2 signaling and plays an important regulatory role in T cells.[89]^22 Analysis of STAT5 CA overexpression CUT&RUN-seq and ATAC-seq data[90]^23 revealed enhanced chromatin accessibility and transcriptional binding at the STS2 locus ([91]figure 1J), suggesting that IL-2-mediated STAT5 activation may contribute to STS2 upregulation in CD8^+ T cells. Collectively, these results demonstrate that STS2 expression in the TME progressively increases with CD8^+ T-cell exhaustion, reaching its highest levels in terminally exhausted CD8^+ T cells. STS2 deficiency enhanced the antitumor activity and immunotherapy effect Analysis using the Tumor Immune Dysfunction and Exclusion score,[92]^24 25 which assesses gene function in T cells, revealed that patients with high CTL (cytotoxic T lymphocyte) infiltration in the STS2 high-expression group had poorer prognosis than in the STS2 low-expression group, suggesting STS2 association with T-cell dysfunction ([93]online supplemental figure S2E,F). Given the high expression of STS2 in CD8^+ terminally Tex cells and its association with T-cell inactivation, we hypothesized that STS2 loss might contribute to improved tumor control. To test this hypothesis, we generated STS2 knockout mice ([94]online supplemental figure S3A–C) and confirmed that STS2 deficiency did not affect lymphocyte numbers or CD3^+ cell numbers and CD4/CD8 cell proportions in peripheral blood and spleen ([95]online supplemental figure S3D–F). In vitro co-culture experiments with MC38 tumor cells and lymphocytes demonstrated that STS2-deficient (STS2^−/−) CD8^+ T cells induced higher rates of late-stage and overall apoptosis in tumor cells ([96]figure 2A and [97]online supplemental figure S3G). To investigate the immune-mediated role of STS2 in tumor control in vivo, we subcutaneously injected MC38 colorectal carcinoma cells into wild-type (WT) and STS2^−/− mice. STS2^−/− mice exhibited enhanced tumor control compared with WT mice ([98]figure 2B). This enhanced tumor control in STS2^−/− mice was also observed in MMTV-PyMT breast tumors ([99]figure 2C) and minimally immunogenic B16F10 melanomas ([100]figure 2D), indicating that STS2 deficiency enables control of diverse tumor types. Figure 2. STS2 deficiency enhanced the antitumor activity and immunotherapy effect. (A) Apoptosis of MC38 tumor cells co-cultured with WT or STS2^−/− CD8^+ T cells at a 3:1 effector:target ratio for 3 hours, measured by annexin V and 7-AAD staining. The CD8^+ T cells were pre-activated in vitro for 3 days with anti-CD3/CD28 and IL-2. n=3 biologically independent samples. Statistical analysis was done by Student’s t-test. (B–C) Temporal analysis of tumor progression (mean size±SEM) in wild-type (WT) and STS2^−/− hosts bearing MC38 (B, n=5) or MMTV-PyMT (C, n=7) neoplasms. Statistical analysis was done by two-way ANOVA. (D) Comparative B16F10 melanoma burden between WT and STS2^−/− mice received B16F10 tumor initiation on day 7 and day 14. n=8 mice per group. Statistical analysis was done by Student’s t-test. (E) MC38 tumor progression dynamics in WT or STS2^−/− mice that received isotype control or anti-CD8-depleting antibody. n=6 mice per group. Statistical analysis was done by two-way ANOVA. (F) MC38 tumor size after NOD/Shi-Prkdc^scid Il2rg^em1/Cyagen mice received 2×10^6 pre-activated WT or STS2^−/− CD8^+T cells (intravenous) on day 8 after tumor initiation. n=8 mice for WT and STS2^−/− groups and 5 mice for no transfer group. Statistical analysis was done by two-way ANOVA. (G–H) Tumor growth in WT or STS2^−/− mice with MC38 (G) and B16F10 (H) that were treated with isotype or anti-programmed cell death protein 1 blocking antibody beginning on day 15 (G) or day 6 (H) after tumor implantation. n=8 mice for MC38 groups and 7 mice for B16F10 groups. Statistical analysis was done by two-way ANOVA. (I) Significantly higher expression of STS2 in CD8^+ T cells from Post versus Pre-ICB treatment at NR patients with melanoma in [101]GSE120575. Statistical analysis was done by Student’s t-test. 7-AAD, 7-aminoactinomycin D; ANOVA, analysis of variance; ICB, immune checkpoint blockade; NR, non-responder; Pre, pre-ICB; Post, post-ICB treatment; R, responder; STS2, suppressor of T-cell receptor signaling 2. [102]Figure 2 [103]Open in a new tab To identify the specific cell type mediating tumor control in STS2^−/− mice, we depleted CD8^+ T cells prior to MC38 tumor initiation. This depletion abolished the tumor control observed in isotype antibody-treated STS2^−/− mice, underscoring the crucial role of CD8^+ T cells in this phenotype ([104]figure 2E and [105]online supplemental figure S3H). To further validate that STS2-mediated tumor control is CD8^+ T cell-intrinsic, we employed an adoptive transfer model. CD8^+ T cells from either WT or STS2^−/− mice were intravenously transferred into NOD/Shi-Prkdc^scid Il2rg^em1/Cyagen mice (which lack mature T, B, and natural killer immune cells) bearing MC38 tumors. The results demonstrated that STS2^−/− CD8^+ T cells exhibited enhanced tumor growth suppression compared with WT CD8^+ T cells ([106]figure 2F and [107]online supplemental figure S3I). The improved CD8^+ T cell-mediated tumor control in STS2^−/− mice suggested that these cells might be more responsive to CD8^+ T cell-enhancing immunotherapies. To test this hypothesis, we treated mice bearing established MC38 and B16F10 tumors with anti-PD-1 blocking antibodies ([108]figure 2G–H and [109]online supplemental figure S3J–K). While B16F10 tumors were better controlled in STS2^−/− mice compared with WT mice, they still progressed ([110]figure 2H and [111]online supplemental figure S2K), allowing for therapeutic intervention to control established tumors. These results indicate that STS2 deficiency in CD8^+ T cells enhanced the efficacy of anti-PD-1 immunotherapy. To validate the antitumor immune and immunotherapy effects of STS2 in patients, we analyzed public datasets. Analysis of single-cell transcriptomics data from patient with melanoma tumor samples treated with checkpoint inhibitors ([112]GSE120575)[113]^26 revealed significant upregulation of STS2 expression in CD8^+ T cells from NR (non-responder) tumors compared with R (responder) tumors following ICB treatment, as well as in post-ICB samples compared with pre-ICB samples ([114]figure 2I). Collectively, these results demonstrate that STS2-deficient CD8^+ T cells contribute to enhanced antitumor immunity and exhibit synergistic effects with immunotherapy. STS2-deficient CD8^+ T cells resist exhaustion and maintain functionality in the TME To elucidate the mechanism by which STS2 deficiency in CD8^+ T cells facilitates long-term tumor control, we analyzed MC38-infiltrating CD8^+ T cells from WT and STS2^−/− mice 12 days post-MC38 implantation using CyTOF based on established methodologies[115]^27 ([116]online supplemental table S1, [117]online supplemental figure S4A). PhenoGraph-based clustering of WT and STS2^−/− CD8^+ T cells yielded eight distinct clusters ([118]figure 3A). In WT CD8^+ T cells, clusters 1 and 2 comprised a larger proportion of WT cells (40% WT vs 17% STS2^−/−), whereas clusters 3 and 4 (24% WT vs 40% STS2^−/−) predominated in STS2^−/− CD8^+ T cells ([119]figure 3B). Protein expression analysis revealed that the dominant WT CD8^+ T-cell clusters (c1 and c2) expressed high levels of TOX, CD103, Lag3, Tim3, CTLA-4, and CD39, the high expression of these exhaustion markers and co-inhibitory receptors indicating a terminally differentiated/exhausted phenotype ([120]figure 3C). Conversely, STS2^−/− CD8^+ T cells showed a higher proportion of TCF1^+PD-1^+CD8^+ T cells (c3) ([121]figure 3C, [122]online supplemental figure S4B), known for their self-renewal capacity and ability to generate terminally differentiated cytotoxic T cells. The predominant clusters in STS2^−/− CD8^+ T cells (c4) expressed lower levels of inhibitory receptors and increased levels of effector-induced protein interferon (IFN)-γ, suggesting an effective and activated state ([123]figure 3C). Notably, the proportions of non-activated, naïve/central memory TCF1^+PD-1^−CD39^− cells (c7) were comparable between WT and STS2^−/− CD8^+ TILs, indicating that STS2 deficiency does not deplete the TCF1^+ stem-like population. Figure 3. STS2-deficient CD8^+T cells resist exhaustion and maintain functionality in the tumor microenvironment. Comprehensive mass cytometry analysis of pooled MC38 tumor-infiltrating CD8^+TILs from WT and STS2^−/− mice (n=5 per genotype). (A–B) High-dimensional visualization (t-SNE) of CD8^+ TIL populations (A) with corresponding cluster frequency analysis (B). (C–D) Expression profiling of functional markers across identified clusters (C) and individual samples (D), presented as normalized Z-scores. (E–G) Analysis of human endometrial cancer specimens (n=3), stratified by STS2 expression levels, including cluster identification (E), frequency distribution (F), and protein expression patterns (G). (H–I) Flow cytometric quantification of CD39^+PD-1^+ (n=5) or PD-1^+ (n=5) (H) or IFN-γ^+ (n=4) (I) of CD8^+ T cells in MC38 tumor. Statistical analysis was done by Student’s t-test. IFN, interferon; PBMC, peripheral blood mononuclear cells; PD-1, programmed cell death protein 1; STS2, suppressor of T-cell receptor; t-SNE, t-distributed stochastic neighbor embedding; tdLN, tumor-draining lymph nodes; TILs, tumor-infiltrating lymphocytes; WT, wild-type. [124]Figure 3 [125]Open in a new tab We also analyzed CD8^+ T cells from peripheral blood and tumor-draining lymph nodes (tdLN). t-SNE (t-distributed stochastic neighbor embedding) dimensionality reduction clustering showed that CD8^+ T cells in peripheral blood and tdLN clustered together and were distinct from tumor-infiltrating CD8^+ T cells ([126]online supplemental figure S4C). Protein expression analysis revealed higher IFN-γ expression in STS2^–/– CD8^+ T cells across all three compartments, PBMC (peripheral blood mononuclear cells), tdLN, and tumor ([127]figure 3D). In tdLN, CD62L and Tcf1 were higher in the STS2^–/– group, while in tumors, expression of immunosuppressive receptors (CTLA-4, PD-1, CD39, Tim3, and Lag3) and Tox was higher in the WT group ([128]figure 3D). To investigate the relationship between STS2 expression and cellular activation in human tumor samples, we analyzed STS2 distribution in CD8^+ TILs from three human endometrial tumor tissues. Clustering CD8^+ TILs based on the upper and lower thirds of STS2 expression revealed distinct enrichment patterns ([129]figure 3E,F, [130]online supplemental figure S4C). Compared with the STS2-high group, cluster 3 showed the largest decrease in STS2-low cells, while cluster 4 showed the largest increase ([131]online supplemental figure S4D–F). Consistently, increased levels of exhaustion markers and co-inhibitory receptors (Tox, PD-1, ICOS, Tim3, Lag3, CTLA4) were observed in the STS2-high subset, whereas activation marker (CD25) was increased in the STS2-low subset in human endometrial tumor tissues ([132]figure 3G). We used spectral flow cytometry to verify the decreased proportion of CD39^+PD-1^+ and PD-1^+CD8^+ cells ([133]figure 3H), and the increased proportion of IFN-γ^+CD8^+ cells ([134]figure 3I) in STS2^–/– mice MC38 tumors ([135]online supplemental figure S4G,H). To further investigate whether STS2 deficiency affects the functional trajectory of CD8^+ T cells under persistent stimulation in vitro, we conducted a temporal analysis of CD8^+ T cells stimulated with anti-CD3/CD28 and IL-2 over 10 days. CyTOF analysis revealed that STS2^–/– CD8^+ T cells maintained higher expression of the effector cytokine IFN-γ at day 10 compared with WT cells ([136]online supplemental figure S5). Similarly, STS2^–/– cells sustained higher levels of proliferation marker Ki67 and activation marker CD25 by day 10, while expressing lower levels of exhaustion-associated markers PD-1 and CD39 ([137]online supplemental figure S5). These in vitro findings support our in vivo observations that STS2 deficiency delays functional decline and acquisition of exhaustion markers under persistent stimulation. In summary, both mouse and human CD8^+ TILs demonstrated that STS2 high-expressing cells were enriched for co-expression of exhaustion markers and co-inhibitory receptors, with STS2 most highly expressed in the most terminally exhausted subsets. STS2-deficient CD8^+ TILs exhibited resistance to exhaustion and maintained functionality within the TME. Transcriptional profiling revealed STS2-deficient CD8^+TILs sustained effector programming and resistance to exhaustion Single-cell transcriptomic data revealed no significant differences in T-cell states between the spleens of tumor-bearing WT and STS2^–/– mice ([138]online supplemental figure S6A). To decipher how STS2 transcriptionally programs CD8^+ TILs, we performed combined scRNA-seq and TCR sequencing plus antibody staining on CD45-enriched tumor-infiltrating cells from WT and STS2^–/– mice, with two samples per group ([139]online supplemental figure S6B,C). Consistent with [140]figure 1, STS2 expression in the immune microenvironment was predominantly in CD8^+ T cells ([141]online supplemental figure S6C,D). Seurat-based clustering of the WT and STS2^–/– CD8^+ TILs resolved six distinct clusters ([142]figure 4A, [143]online supplemental table S2), with notable differences in cluster proportions between the two groups ([144]figure 4B, [145]online supplemental figure S6E). Figure 4. STS2-deficient CD8^+TILs sustained effector programming and resistance to exhaustion. (A–B) Single-cell transcriptomic profiling of WT and STS2^–/– CD8^+TILs (A) with quantitative distribution analysis across identified clusters (B). (C) Bubble plot showing relative expression of cell-state-associated genes. Bubble size is proportional to the percentage of cells expressing a gene and color intensity is proportional to average scaled gene expression. (D) Heatmap illustrating expression of 19 curated gene signatures across CD8^+TILs cluster. Heatmap was generated based on the scaled gene signature scores. (E) The terminal exhausted signature score of WT and STS2^–/– CD8^+TILs. Statistical analysis was done by Student’s t-test. The trend lines connected the median values between groups. (F) RNA velocity analysis from WT and STS2^–/– CD8^+TILs in figure 4A for clusters 1–6. Arrows show directions of cell state transitions. (G) Pseudotime analysis of the indicated states from [146]online supplemental figure S4G. (H) The single cell counts of each clone type in each cluster. (I) Frequency of CD8^+TILs cells within paired TCR sequences. Clonotype frequency (X) indicates the relative abundance of each T-cell clone within the analyzable TCR repertoire: medium (1–10%), large (10–100%), and hyper-expanded (>100%, theoretical maximum). The frequency (X) represents the proportion of cells belonging to each clonotype among the CD8^+ TILs that have paired TCR sequences. IFN, interferon; STS2, suppressor of T-cell receptor signaling 2; TCR, T-cell receptor; TILs, tumor-infiltrating lymphocytes; UMAP, uniform manifold approximation and projection; WT, wild-type. [147]Figure 4 [148]Open in a new tab The naïve-like cluster (c1) expressed stemness-associated markers Tcf7 (encoding TCF1), Sell (encoding CD62L), and Slamf6 (encoding Ly108) and displayed a naïve-like phenotype with high expression of a naïve gene signature ([149]online supplemental table S3), and its abundance was comparable between WT and STS2^−/− cells (15% WT vs 13% STS2^−/−) ([150]figure 4B–D). Notably, cluster 2 (IFN−responsive effector memory), which highly expressed the effector-associated chemokine Ccl5, effector memory marker Ly6c2, and signatures for IFN response (Isg15, Bst2, Ifi27l2a), cytotoxicity, and TCR signaling, along with the activated cluster (c3) expressing NF-κB signaling pathway gene Nfkbia, AP-1 family member Junb, NR4A family transcription factor Nr4a1, and survival factor receptor Il7r, together accounted for 39% of STS2^−/− CD8^+ TILs but only 18% in WT CD8^+ TILs ([151]figure 4B–D). Clusters 4, 5, and 6 expressed Tox, a key regulator of exhaustion, and showed lower proportions in the STS2^−/− group compared with WT (47% vs 68% total) ([152]figure 4B,C). Among these, the metabolically active cluster (c4) exhibited high expression of glycolysis (Ldha), DNA replication (MCM family), and protein synthesis (Eif5a) genes, along with elevated oxidative phosphorylation and glycolysis signatures. The proliferating cluster (c5) exhibited high expression of cell cycle-related markers (Mki67, Top2a, Stmn1) and showed the MAPK signaling signature. Clusters 6 showed abundant expression of Pdcd1 (encoding PD-1) and Havcr2 (encoding TIM3), with the highest expression of co-inhibitory receptors Entpd1 (encoding CD39), Cd38, and Cd244a ([153]figure 4B,C). Remarkably, STS2^−/− CD8^+ TILs downregulated terminally exhausted gene signatures ([154]online supplemental table S3), while maintaining progenitor and intermediate exhausted gene signatures[155]^21 ([156]figure 4E and [157]online supplemental figure S6F). To infer directionality of CD8^+T cell development, we assessed RNA velocity[158]^28 overlaid on the UMAP (uniform manifold approximation and projection) of cells from all identified clusters ([159]figure 4F). We identified continuous trajectories aligning CD8^+ T-cell subpopulations, particularly evident from clusters 2 and 3 to 6, supporting differentiation from activated and/or IFN−responsive effector memory cells to terminal exhausted cells ([160]figure 4F, up). Consistently, this trajectory was observed when visualizing WT or STS2^−/− cell clusters separately ([161]figure 4F, down). To further understand the differentiation trajectories of CD8^+ T cells, we projected the expression of naïve, activation/effector, cytotoxicity, and exhaustion genes and signatures onto the UMAP ([162]online supplemental figure S6G,H). Building on this, Monocle3 analysis revealed the differentiation trajectories ([163]online supplemental figure S6I), and pseudotime analysis indicated that STS2^−/− cells mainly accumulated in the intermediate differentiation cell state ([164]figure 4G), further supporting the idea that STS2 deficiency delays terminal differentiation. Single-cell TCR sequencing provided insights into the clonal dynamics of tumor-infiltrating T cells ([165]online supplemental figure S6J). We observed hyper-expanded clones were predominantly enriched in the terminal exhausted CD8^+ T-cell cluster, suggesting that extensive proliferation may be associated with T-cell dysfunction rather than enhanced antitumor activity ([166]figure 4H). Moreover, we analyzed the expression of effector function, cytotoxicity, and TCR signaling signatures across different clone size categories (medium, large, and hyper-expanded), which revealed that hyper-expanded clones exhibited lower signature scores for these functional pathways compared with medium and large clones ([167]online supplemental figure S6K). Furthermore, the proportion of hyper-expanded clones was lower in STS2^−/− mice compared with WT mice (17% vs 23%) ([168]figure 4I), suggesting that STS2 deficiency inhibits T-cell differentiation toward a highly expanded but functionally compromised state under persistent antigen stimulation. Finally, to validate our analysis in the context of established T-cell states, we used the R package ProjecTILs[169]^17 to project CD8^+ TILs onto the mouse CD8^+ TIL reference atlas. Consistent with our previous observations, the proportion of CD8_Tex was higher in the WT group, while the proportion of effector memory CD8^+ T cells was higher in STS2^−/− cells ([170]online supplemental figure S6L). These findings collectively demonstrate that STS2 deficiency promotes a more functional CD8^+ T-cell repertoire within the TME, characterized by sustained effector programming and resistance to terminal exhaustion. STS2 broadly regulates T-cell function and modulates PD-1 expression at the post-transcriptional level To investigate the regulatory mechanisms of STS2 in lymphocytes, we performed RNA sequencing (RNA-seq) and ATAC-seq (assay for transposase-accessible chromatin using sequencing) on lymphocytes isolated from the spleens of WT and STS2^−/− mice ([171]figure 5A–D), which were stimulated in vitro for 3 days with anti-CD3/CD28 and IL-2. The RNA-seq results revealed significant changes at the mRNA level in lymphocytes due to STS2 deficiency. Among the most significantly downregulated genes in the STS2^−/− group were stress-related heat shock genes (such as HSPA1A and HSPA1B) ([172]figure 5A), which have been implicated in resistance to immunotherapy.[173]^29 In terms of immune regulatory pathways, STS2 deficiency led to transcriptional changes in the expression of surface molecules (Btla, Ctla4, Cd247, Lag3, Cd3e), cytokines (Il4, Il5, Il10, Ccl3, Ccl4), effector molecules (Tnf, Ifng, Gzmb, Prf1), and transcription factors (Tox, Jun, Tbx21) ([174]figure 5B), as well as alterations in their chromatin accessibility ([175]figure 5D), indicating that the TCR and CD28 pathways were affected by the absence of STS2. Pathway enrichment analysis of differentially expressed genes showed that the genes regulated by STS2 were primarily enriched in processes related to the inflammatory response, regulation of intracellular signal transduction, and regulation of protein metabolic processes ([176]figure 5C), consistent with previous reports.[177]1030,[178]32 Figure 5. STS2 broadly regulates T-cell function. (A) Volcano plot showing DEGs in RNA-seq of WT and STS2^−/− lymphocytes stimulated in vitro for 3 days with anti-CD3/CD28 and IL-2. (B) Heatmap showing relative expression (Z score of the normalized counts) of surface molecules, cytokines and effector molecules and transcriptional factors in RNA-seq of WT and STS2^−/− lymphocytes. (C) Bubble plot of GO pathways enriched in DEGs between WT and STS2^−/− lymphocytes. The size of the dots represents the counts of enriched genes, and the color represents the enrichment score. (D) ATAC-seq tracks of the indicated gene locus in activated WT and STS2^−/− lymphocytes. (E) Bubble plot of GO pathways enriched in STS2-interacting proteins. The size of the dots represents the counts of enriched genes, and the color represents the enrichment score. (F) Apoptosis of WT or STS2^−/− lymphocytes co-cultured with MC38 tumor cells measured by annexin V and propidium iodide (PI) staining. n=3 biologically independent samples. Statistical analysis was done by Student’s t-test. (G) In vitro proliferation of CD8^+T cells from WT and STS2^−/− mice stimulated by anti-CD3, anti-CD28 and IL-2 for 4 days and analyzed by CellTiter 96 AQueous One Solution Cell Proliferation Assay. Statistical analysis was done by two-way analysis of variance. (H) RT-qPCR analysis of Mki67 expression in WT and STS2^−/− CD8^+ TILs. Statistical analysis was done by Student’s t-test. (I) Co-IP detection of the interaction between STS2 with CD3ε and c-Cbl. ATAC-seq, assay for transposase-accessible chromatin using sequencing; Co-IP, co-immunoprecipitation; DEG, differentially expressed gene; GO, gene ontology; IL, interleukin; mRNA, messenger RNA; RNA-seq, RNA sequencing; RT-qPCR, reverse transcription quantitative polymerase chain reaction; STS2, suppressor of T-cell receptor signaling 2; WT, wild-type. [179]Figure 5 [180]Open in a new tab To further explore the regulatory mechanism of STS2 in lymphocytes, we analyzed the interacting proteins of STS2 reported by a previous study[181]^10 ([182]figure 5E). Interestingly, proteins interacting with STS2 were enriched in the regulation of cell death pathways. This led us to investigate the impact of STS2 on lymphocyte apoptosis. After co-culturing lymphocytes and MC38 tumor cells in vitro, we observed that both late and total apoptosis in the STS2^−/− lymphocytes were significantly lower than those in the control group ([183]figure 5F and [184]online supplemental figure S7A). Additionally, we found that the proliferation rate of STS2-deficient lymphocytes in vitro was significantly higher than that of the WT group ([185]figure 5G), as well as the level of Mki67 mRNA expression was higher in STS2^−/− CD8^+TILs ([186]figure 5H). Previous studies have demonstrated that STS2 regulates T-cell function through multiple pathways, particularly impacting TCR signaling.[187]^7 33 34 Unlike STS1, another member of the STS family, STS2 exerts its influence not through phosphatase activity, but rather by interacting with key nodes in the TCR pathway.[188]35,[189]37 To further elucidate this mechanism, we performed in vitro experiments to detect the phosphorylation of Zap70 and Erk1/2, key nodes in the TCR signaling process, following anti-CD3/CD28 stimulation. There was no significant difference between the STS2^−/− and WT groups ([190]online supplemental figure S7B). Additionally, we confirmed the interaction of STS2 with Cbl-b and CD3ε through co-immunoprecipitation assays ([191]figure 5I). Notably, the transcriptional level of Pdcd1 was increased in STS2^−/− CD8^+TILs ([192]figure 6A, right), but cell-surface levels of PD-1 were downregulated in STS2^−/− CD8^+T cells in tumors ([193]figure 3H), peripheral blood, and spleen ([194]figure 6A, left). We speculate that regulation at the transcriptional level may be due to the interaction of STS2 with the IκB complex (TAK1 and NEMO),[195]^30 which inhibits TCR-induced NFκB signaling through ubiquitin-dependent inhibition, thereby suppressing PD-1 transcription. STS2’s interacting proteins and regulated genes were enriched in pathways related to protein localization, synthesis, and degradation, such as cellular protein localization, cellular component biogenesis, and regulation of protein metabolic processes ([196]figure 5C,E). Given that STS2 is involved in the regulation of protein metabolic processes, we hypothesize that STS2 may interact with PD-1 at the protein level. Co-immunoprecipitation experiments confirmed the interaction between ectopically expressed Flag-tagged PD-1 and Myc-STS2 in 293T cells ([197]figure 6B), and endogenously in Jurkat cells ([198]figure 6C). These data suggest that STS2 interacts with PD-1 and mediates its downregulation by protein metabolic processes. Figure 6. STS2 modulates PD-1 expression at the post-transcriptional level. (A) The ratio of PD-1^+CD8^+ of CD8^+ cells in PBMC and spleen cells from MC38 tumor-bearing mice in WT and STS2^−/− mice (left). RT-qPCR analysis of Pdcd1 expression in WT and STS2^−/− CD8^+ tumor-infiltrating lymphocytes (right). Statistical analysis was done by Student’s t-test. (B–C) Validation of the interaction between STS2 and PD-1 by Co-IP in 293T (B) and Jurkat cells (C). (D) Immunoblotting showing the construction of STS2 knockout cells in Jurkat cells. (E–H) Change in the levels of PD-1 on the cell surface after treatment with the proteasome inhibitor MG132 (E–F) or the lysosome inhibitor CQ (G–H) in Jurkat (E, G) or mice lymphocytes (F, H). n=4 for Jurkat cells per group and n=3 for mice lymphocytes per group. Statistical analysis was done by Student’s t-test. (I) GSEA plots showing enrichment in PROTEASOME and ENDOCYTOSIS pathways of DEGs between WT and STS2^−/− lymphocytes RNA-seq. (J–K) The remaining antibody-labeled surface PD-1 was detected with an APC-conjugated secondary antibody in Jurkat (J) or mice lymphocytes (K). Statistical analysis was done by two-way analysis of variance. Co-IP, co-immunoprecipitation; GSEA, gene set enrichment analysis; MFI, mean fluorescent intensity; NES, normalized enrichment score; PBMC, peripheral blood mononuclear cells; PD-1, programmed cell death protein 1; RNA-seq, RNA sequencing; RT-qPCR, reverse transcription quantitative polymerase chain reaction; STS2, suppressor of T-cell receptor signaling 2 ;t-SNE, t-distributed stochastic neighbor embedding; WT, wild-type. [199]Figure 6 [200]Open in a new tab To elucidate the mechanism of STS2-mediated PD-1 regulation, we generated STS2-knockout (sgSTS2) Jurkat cells ([201]figure 6D) and examined whether STS2 affects PD-1 degradation via the proteasome or lysosome pathways. Treatment with MG132, a specific proteasome inhibitor, protected the PD-1 protein from degradation in both sgSTS2 Jurkat cells and mouse STS2^–/– lymphocytes ([202]figure 6E,F), suggesting the involvement of the proteasome in STS2 regulation, which is consistent with the enrichment in the proteasome pathway of DEGs (differentially expressed genes) in RNA-seq of STS2^–/– lymphocytes ([203]figure 6I, top). Moreover, we found that knockdown of STS2 caused increased PD-1 ubiquitination in Jurkat cells ([204]online supplemental figure S7C). Interestingly, lysosome inhibitors also protected PD-1 protein from degradation in these cells ([205]figure 6G,H), indicating that both proteasomal and lysosomal pathways may be involved in STS2-mediated PD-1 regulation. Given that STS2 can affect the endocytosis pathway ([206]figure 6I, down), as demonstrated by its ability to facilitate the endocytosis of cell-surface TCR–CD3 complexes and/or inhibit their recycling back to the plasma membrane,[207]^10 we explored whether STS2 influences PD-1 endocytosis. We employed a surface labeling assay to track PD-1 internalization ([208]online supplemental figure S7D). Surprisingly, our results showed that in the STS2-deficient group, although the initial PD-1 content on the membrane surface was relatively low, the proportion of PD-1 remaining on the membrane surface after endocytosis was higher compared with the control group ([209]figure 6J,K). This suggests that STS2 deficiency inhibits the endocytosis of PD-1, potentially contributing to its altered expression and function in STS2^–/– T cells. Collectively, these findings demonstrate that STS2 plays a multifaceted role in regulating T-cell function, including modulation of cell death, proliferation, and protein metabolic process. Importantly, our results reveal a novel mechanism by which STS2 regulates PD-1 expression through both transcriptional and post-transcriptional processes, including protein–protein interactions, degradation pathways, and endocytosis. This intricate regulation of PD-1 by STS2 may contribute to the enhanced antitumor immunity observed in STS2-deficient T cells. Discussion In this study, we have uncovered a critical role for STS2 in regulating CD8^+ T-cell exhaustion and antitumor immunity. Our findings reveal that STS2 deficiency enhances T cell-mediated tumor control and improves the efficacy of checkpoint blockade immunotherapy ([210]figure 7). This effect is mediated through multiple mechanisms, including the maintenance of effector function, resistance to terminal exhaustion, and modulation of PD-1 expression. Figure 7. STS2 deficiency enhances T cell-mediated tumor control and improves the efficacy of checkpoint blockade immunotherapy. ICB, immune checkpoint blockade; PD-1, programmed cell death protein 1; STS2, suppressor of T-cell receptor signaling 2; WT, wild-type. [211]Figure 7 [212]Open in a new tab The upregulation of STS2 in tumor-infiltrating CD8^+ Tex cells, as demonstrated by our scRNA-seq and protein-level analyses, suggests that STS2 may serve as a marker of T-cell exhaustion in the TME. This observation is consistent across multiple tumor types and species, indicating a conserved role for STS2 in T-cell biology. The gradual increase in STS2 expression with T-cell exhaustion progression implies that STS2 may be involved in the transition from effector to exhausted states. Our transcriptional profiling revealed that STS2 deficiency promotes a more functional CD8^+ T-cell state within tumors. The increased proportion of activated and effector-like cells and the maintenance of IFN-responsive states in STS2^−/− CD8^+ TILs suggest that STS2 may act as a molecular brake on T-cell function. By removing this brake, STS2-deficient T cells resist terminal exhaustion and maintain antitumor potential through enhanced TCR signaling and decreased PD-1 expression. This finding aligns with the current understanding of T-cell exhaustion as a plastic manipulable for therapeutic benefit.[213]^38 39 The broad regulatory effects of STS2 on T-cell function, including its impact on cell death, proliferation, and protein metabolic process, highlight its potential as a multifaceted regulator of T-cell responses. STS2 enhances endocytosis and downregulation of TCR-CD3 complexes after TCR stimulation in Jurkat cells.[214]^10 Moreover, STS2 inhibits TCR signaling by interacting with c-Cbl and Cbl-b of the Cbl family.[215]^40 41 Our discovery of STS2’s interaction with PD-1 and its involvement in PD-1 regulation at both transcriptional and post-transcriptional levels is particularly intriguing. Notably, our results suggest STS2’s role in regulating PD-1 through a ubiquitin-dependent mechanism. This finding is consistent with previous studies showing that STS2 interacts with EGFR and regulates its expression through Cbl-mediated ubiquitination and subsequent degradation,[216]^41 and that Nrdp1 promotes Zap70 ubiquitin-dependent degradation in an STS2-dependent manner, thereby negatively regulating TCR signaling pathways.[217]^42 Interestingly, this finding aligns with recent studies on post-translational modifications of PD-1. For instance, TOX has been shown to affect the endocytic recycling pathway of PD-1,[218]^14 FBXO38 can influence PD-1 ubiquitination,[219]^15 and UFL1 affects the UFMylation of PD-1.[220]^43 Our discovery that STS2 can affect PD-1 post-translational modification through multiple pathways adds to this growing body of evidence and underscores the complex regulation of this crucial immune checkpoint molecule. From a therapeutic perspective, our results suggest that targeting STS2 could be a promising strategy to enhance antitumor immunity. The synergistic effect observed between STS2 deficiency and anti-PD-1 immunotherapy is particularly encouraging. This combination approach could potentially overcome resistance to checkpoint blockade therapies, a significant challenge in cancer immunotherapy.[221]^44 45 However, several questions remain to be addressed. It is important to note that our study primarily examines polyclonal rather than antigen-specific T-cell responses. Future studies employing antigen-specific models, such as TCR transgenic T cells or defined tumor antigens, would further refine our understanding of how STS2 influences antigen-specific T-cell exhaustion and antitumor immunity. The precise molecular mechanisms by which STS2 promotes T-cell exhaustion and PD-1 expression need further elucidation. Additionally, the potential off-target effects of STS2 inhibition on other immune cell populations and normal tissues should be carefully evaluated. The translational potential of these findings will depend on the development of specific STS2 inhibitors and their evaluation in preclinical models. In conclusion, our study identifies STS2 as a key regulator of CD8^+ T-cell exhaustion and antitumor immunity. By maintaining T-cell functionality, modulating PD-1 expression, and influencing the balance between effector and exhausted states, STS2 deficiency enhances tumor control and improves responses to immunotherapy. These findings not only advance our understanding of T-cell biology in the TME but also pave the way for novel therapeutic strategies in cancer immunotherapy. Future studies should focus on developing targeted approaches to modulate STS2 function in T cells, potentially opening new avenues for enhancing antitumor immune responses in patients with cancer. Materials and methods Cell culture and isolation Cell lines MC38, B16F10, Jurkat, and 293T were obtained from the National Infrastructure of Cell Line Resources (Beijing, China). All cell lines were maintained in Dulbecco’s Modified Eagle Medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. Cells were cultured at 37°C in a humidified incubator with 5% CO[2]. CD8^+ T cells were isolated from mice splenocytes and then purified by negative selection using the EasySep Mouse CD8^+ T Cell Enrichment Kit (STEMCELL) according to the manufacturer’s instructions. Animal studies C57BL/6N mice were purchased from Beijing Vital River Laboratory Animal Technology (Beijing, China). Animals were housed under specific pathogen-free conditions with a 12-hour light/dark cycle and ad libitum access to standard chow and water. All experiments used age-matched and sex-matched mice between 7 and 10 weeks old at the start of each study. Generation of STS2 knockout mice STS2 knockout (STS2^−/−) mice were generated on the C57BL/6N background by Cyagen Biotechnology using CRISPR-Cas9 technology. Exons 2–5 of the STS2 gene were targeted for deletion. Cas9 mRNA and guide RNAs (gRNAs) were co-injected into fertilized eggs. Resulting pups were genotyped by PCR and confirmed by sequencing analysis. The usability of the STS2 antibody in FC (flow cytometry) and CyTOF was validated by staining immune cells derived from WT and STS2^−/− mice ([222]online supplemental figure S1A,B). The following sgRNA sequences for mice STS2 gene knockout were used: gRNA1 (matching forward strand of gene): 5'- TTACTGCTTAAGCCGTAGGGTGG-3'; gRNA2 (matching reverse strand of gene): 5'- TCCCTCATGCTTATTCCCTGTGG-3'. Human tissue samples Surgical specimens were obtained from the patients with endometrial cancer at Beijing Obstetrics and Gynecology Hospital (Approval ID: 2022-KY-037–01). TMAs were constructed by harvesting 400 µm tissue cores from paraffin-wax embedded samples collected from 180 patients with colorectal cancer and 62 patients with endometrial cancer. Statistical analysis Statistical analyses and generation of graphics were performed using GraphPad Prism V.8. All data were expressed as mean±SEM. For statistical results, when only two groups were applied, Student’s t-test was used; when more than two groups were applied, one-way analysis of variance (ANOVA) was used. For the tumor growth data, two-way ANOVA was used. Details of statistical tests used are indicated in the respective figure legends. P value<0.05 (two-tailed) was considered statistically significant, with *p<0.05, **p<0.01, and ***p<0.001, respectively. Other materials and methods were provided in the [223]online supplemental file 2. Supplementary material online supplemental file 1 [224]jitc-13-6-s001.pdf^ (3.2MB, pdf) DOI: 10.1136/jitc-2024-010735 online supplemental file 2 [225]jitc-13-6-s002.pdf^ (285.1KB, pdf) DOI: 10.1136/jitc-2024-010735 online supplemental file 3 [226]jitc-13-6-s003.pdf^ (231.2KB, pdf) DOI: 10.1136/jitc-2024-010735 Acknowledgements