Abstract Metabolic reprogramming of tumor cells dynamically reshapes the distribution of nutrients and signals in the tumor microenvironment (TME), affecting intercellular interactions and resulting in metabolic immune suppression. Increased glucose uptake and metabolism are characteristic of many tumors. Meanwhile, the progression of colorectal carcinoma (CRC) relies on lipid metabolism. Therefore, investigating the role of glucolipid metabolic reprogramming on tumor immunity contributes to identifying new targets for immune suppression intervention in CRC. Our previous work demonstrated that SIRT1 is the hub gene involved in glucolipid metabolic conversion in CRC. Here, it is found that upregulated SIRT1 in CRC cells increases Treg functionality by promoting the secretion of CX3CL1. The CX3CL1‐CX3CR1 signaling activated transcription factors SATB1 and BTG2, promoting the differentiation of TCF7^+ Treg cells into functionally enhanced TNFRSF9^+ Treg cells. Multiplex immunofluorescence (mIHC) analysis of a CRC tissue microarray confirmed the promoting effect of CX3CL1 on Treg infiltration. Additionally, the therapeutic efficacy of CX3CR1 inhibitor monotherapy and combination therapy is validated with the PD‐1 antibody in the humanized subcutaneous CRC mouse model. This study elucidates a potential mechanism that metabolic reprogramming of cancer cells collaborates with subsequent immunosuppression to promote CRC progression. Keywords: Colorectal carcinoma, CX3CL1, Immunosuppression, Regulatory T cell, SIRT1 __________________________________________________________________ This study discovers that SIRT1, a hub gene involved in glucolipid metabolic conversion in colorectal carcinoma (CRC), stimulates CX3CL1 secretion in CRC cells by activating FOXO1. The CX3CL1‐CX3CR1 signaling promotes the differentiation of TCF7^+ regulatory T cells (Tregs) into an enhanced immunosuppressive TNFRSF9^+ Treg phenotype. CRC cells exert metabolic immunosuppression on the tumor microenvironment via the SIRT1‐CX3CL1 axis. graphic file with name ADVS-12-2404734-g006.jpg 1. Introduction Metabolic reprogramming and immune escape are two fundamental hallmarks of cancer. Nevertheless, they are not mutually exclusive. The dynamic variation of nutrients and signals within the TME drives the metabolic plasticity of cancer cells and facilitates the immune suppression of effector cells.^[ [60]^1 ^] In contrast to the traditional Warburg effect, the progression of CRC relies on lipid metabolism.^[ [61]^2 ^] However, whether glucolipid metabolic reprogramming affects the crosstalk between tumor and immune cells is elusive. Our previous studies revealed that the deacetylase SIRT1, which is widely distributed in the cytoplasm and nucleus, is a metabolic switch between glycolysis and fatty acid oxidation, its upregulation enables tumor cells to adapt to glucose deprivation and promotes tumor progression.^[ [62]^3 ^] Given that high SIRT1 expression in tumor cells can affect the expression and secretion of cytokines, thus influencing tumor growth and metastasis, further exploration of whether SIRT1 inhibits tumor immunity is warranted.^[ [63]^4 ^] Chemokines and their receptors have been a hot topic in tumor immunity research. The exocrine cytokine CX3CL1 (C‐X3‐C Motif Chemokine Ligand 1, also known as Fractalkine) has been shown to play an important role in the immune response. It is traditionally believed that CX3CL1 exerts an antitumor effect by recruiting CD8^+ T cells. However, CX3CL1 participates in tumor invasion and metastasis in some tumors, leading to a poor prognosis.^[ [64]^5 ^] Recent studies have indicated that the interaction between tumor cells and other cells in the microenvironment mediated by CX3CL1, such as macrophages and mesenchymal stem cells, is related to developing an immunosuppressive environment.^[ [65]^6 , [66]^7 ^] Therefore, a comprehensive analysis of the mechanism of CX3CL1 production and function contributes to unraveling its effector network in TME. Regulatory T cells (Tregs) are one of the main cell types that exert immunosuppressive effects in the tumor microenvironment. Their infiltration is closely associated with adverse clinical phenotypes such as treatment resistance, immune evasion, and metastasis. Recent studies have shown that tumor cells can recruit Treg cells to exert immunosuppressive effects and promote tumor progression by secreting certain cytokines, such as CCL17 and CCL20.^[ [67]^8 , [68]^9 ^] Notably, with the emergence of single‐cell RNA sequencing (scRNA‐seq) technology, it has become increasingly recognized that Treg cells exhibit heterogeneity in the tumor environment. A pan‐cancer analysis indicated that TNFRSF9^+ Treg (TNF Receptor Superfamily Member 9, also known as 4‐1BB and CD137) cells are widely present in various types of tumor tissues and exert strong immunosuppressive effects.^[ [69]^10 ^] The transcription factor TCF‐1 (encoded by TCF7) plays a crucial role in the development of Treg cells, but in CRC, TCF‐1‐deficient Treg cells exhibit stronger immunosuppressive effects than WT Treg cells.^[ [70]^11 ^] These studies suggest that further elucidating the impact of cytokines on Treg phenotypes and functions is highly important. In this study, we discovered that SIRT1 promotes the upregulation of CX3CL1 in CRC cells. CX3CL1, in turn, enhances the function of Tregs and suppresses antitumor immunity. Our findings suggested that tumor metabolic reprogramming and immune evasion cooperate to promote tumor progression. 2. Results 2.1. SIRT1 is Closely Related to the Immunosuppressive Tumor Microenvironment The rapid energy provision and the production of key precursors for the biosynthesis of macromolecules make glycolysis the preferred energy supply method for cancer cells. However, metabolomic analysis of a large cohort study has found that the progression of CRC was highly dependent on lipid metabolism,^[ [71]^2 ^] suggesting that glucolipid metabolic reprogramming is involved in the development of CRC. To explore the specific roles of glycolysis and lipid metabolism in CRC progression, we conducted a univariate Cox regression analysis of all glycolysis‐ and lipid metabolism‐related genes in CRC samples from the TCGA database (n = 392). We identified 126 genes that were significantly associated with poor or prolonged survival in CRC (univariate Cox p < 0.05). The expression matrix of these genes served as the input for consensus clustering. Four groups were identified by consensus clustering: C1, C2, C3, and C4 (Figure [72] 1A). Kaplan‒Meier (K‒M) survival analysis indicated that the prognosis of the C4 group was significantly worse than that of the other groups (Figure [73]1B). Subsequently, we performed gene set enrichment analysis (GSEA) of the differentially expressed genes (DEGs) between patients in the C4 group and those in the C1 group. The results revealed a significant enrichment of lipid metabolism‐related gene sets and a markedly decreased enrichment of effector immune response‐related gene sets. At the same time, changes in glycolysis‐related pathways were not prominent (Figures [74]1C,D and [75]S1A, Supporting Information). Correspondingly, we observed increased enrichment of pathways associated with macrophage differentiation and regulatory T‐cell differentiation related to immune suppression in patients in the C4 group (Figure [76]S1B,C, Supporting Information). These results indicated that the enhanced lipid metabolism resulting from glucolipid metabolic reprogramming might promote the progression of CRC by reshaping the tumor immune microenvironment (TIME). Figure 1. Figure 1 [77]Open in a new tab SIRT1 is closely related to the immunosuppressive tumor microenvironment. A) Consensus clustering of glucolipid metabolism‐related genes, which are significantly associated with prognosis, identifying four groups in the TCGA colorectal cancer cohort (TCGA‐COAD, n = 392). B) Kaplan‐Meier estimates of survival for patients with different glucolipid subtypes in the TCGA‐COAD cohort. C) The enrichment score of the “Lipid metabolic process” in C4 patients versus C1 patients was analyzed by GSEA using RNA‐seq data from the TCGA‐COAD cohort. D) Enrichment score of the “Immune effector process” in C4 patients versus C1 patients, analyzed by GSEA using RNA‐seq data of TCGA‐COAD cohort. E) The correlation of 10 main tumor‐infiltrating cell types with SIRT1 in CRC tissues. Red: positive correlation; blue: negative correlation. F) Representative gross appearance (left) of the subcutaneous allografts in the indicated groups. Tumor growth curves (right) of C57BL/6J mice inoculated with Sirt1‐KO or control MC38 allografts. Statistical significance was assessed by student t‐test of variance. ***p < 0.001. G) Flow cytometry analysis of the ratio of Cd206^+ cells in Cd11b^+ cells. n = 7, student t‐test of variance, ***p < 0.001. H) Flow cytometry analysis of the ratio of Cd25^+Cd127^− cells in Cd4^+ T cells. n = 7, student t‐test of variance, ***p < 0.001. I–K) Flow cytometry analysis of the ratio of Pd‐1^+ cells in Cd8^+ T cells (I), Tim‐3^+ cells in Cd8^+ T cells (J), and Ctla4^+ cells in Cd8^+ T cells (K). n = 7, student t‐test of variance, ***p < 0.001. L) Elisa of effector cytokines IFNγ (left), TNFα (right) from Sirt1‐KO or control MC38 allografts. n = 7, student t‐test of variance, ***p < 0.001. Our previous study revealed that SIRT1 was the hub that facilitated the metabolic shift from glycolysis to lipid metabolism in CRC cells.^[ [78]^3 ^] Here, we assessed whether SIRT1 shaped the tumor immune environment. First, we examined the correlation between SIRT1 expression and the levels of tumor‐infiltrating immune cells using the CIBERSORT algorithm.^[ [79]^12 ^] We found that SIRT1 was positively correlated with the infiltration of regulatory T cells (Tregs), but negatively correlated with CD8^+ T cells (Figure [80]1E). Moreover, we discovered a positive correlation between SIRT1 mRNA expression and the expression of T‐cell exhaustion signature genes in CRC samples by performing Gene Expression Profiling Interactive Analysis (GEPIA) (Figure [81]S1D, Supporting Information). Moreover, SIRT1 expression was positively correlated with markers of M2 macrophages and Tregs (Figure [82]S1E,F, Supporting Information). These results suggested that SIRT1 participated in shaping the immunosuppressive tumor microenvironment. Next, we established Sirt1 knockout (Sirt1‐KO) MC38 mouse CRC cell lines using lentiviral vectors and confirmed gene knockout efficiency by western blot analysis (Figure [83]S1G, Supporting Information). Next, we constructed a subcutaneous allograft tumor model in C57BL/6 mice and generated tumor growth curves for the wild‐type (WT) MC38 and MC38‐Sirt1KO groups. When the living tumor volume in any tumor‐bearing mice exceeded 2000 mm^3, all the mice were euthanized, and the tumor tissues were harvested and weighed. The experimental results indicated a significant inhibition of tumor progression in the Sirt1‐KO group (Figure [84]1F). Furthermore, we processed all tumor tissues by digesting them into single cells and then analyzed the infiltration of immune cells using flow cytometry. The results showed that the Sirt1‐KO group exhibited a significant decrease in the infiltration of immunosuppressive cells, such as M2‐type macrophages (Cd11b^+Cd206^+) and Treg cells (Cd4^+Cd25^+Cd127^−), within the tumor tissue (Figure [85]1G,H). Meanwhile, we found that the expression level of SIRT1 did not affect the infiltration of myeloid‐derived suppressor cells (MDSCs, Cd11b^+Gr1^+) and dendritic cells (DCs, Cd11c^+Cd86^+) (Figure [86]S1H,I, Supporting Information). Moreover, the expression of exhaustion‐related immune checkpoint markers (Pd‐1, Tim‐3, and Ctla4) was notably reduced in CD8^+ T cells within the Sirt1‐KO cohort compared to the control group (Figure [87]1I–K). Correspondingly, we detected significantly increased levels of IFN‐γ and TNF‐α in the supernatants of the Sirt1‐KO single‐cell cultures (Figure [88]1L). These findings were consistent with previous bioinformatics analysis results, indicating that SIRT1 plays a crucial role in shaping the immunosuppressive tumor microenvironment. 2.2. SIRT1 Promotes the Expression of CX3CL1 in CRC Cells We subsequently evaluated the impact of alterations in SIRT1 expression levels within tumor cells on tumor immunity. Previous studies have highlighted the pivotal role of chemokines in facilitating the interplay between tumor cells and immune cells, orchestrating the generation and recruitment of immune cells that foster a tumor‐promoting microenvironment.^[ [89]^13 ^] Therefore, we hypothesized that SIRT1 might be associated with the secretion of certain chemokines. To test this hypothesis, we first constructed overexpression and knockdown plasmids for SIRT1, and transduced them into two human CRC cell lines, HCT116 and SW480, to obtain cell models with either SIRT1 overexpression (OE) or SIRT1 knockdown (KD). Subsequently, we isolated T cells from human peripheral blood mononuclear cells (PBMCs) and activated them with anti‐CD3, anti‐CD28 antibodies, and IL‐2 for 72 h. Then, we seeded tumor cells in the basolateral chamber of a transwell system and added the activated T cells to the apical chamber (Figure [90]S2A, Supporting Information). The coculture mixture was maintained for 72 h, after which cell culture supernatants were collected to assess the effector cytokines IFN‐γ and TNF‐α expression in the different groups (Figures [91] 2A,B and [92]S2B,C, Supporting Information). We also evaluated the expression levels of PD1 and Ki67 in CD8^+T cells to assess their exhaustion state (Figures [93]2C,D and [94]S2C,D, Supporting Information). Compared to those cultured with tumor cells from the control group, T cells cocultured with tumor cells from the SIRT1‐OE group exhibited reduced IFN‐γ and TNF‐α production, decreased Ki‐67 expression, and increased PD‐1 expression. Coculture of T cells with SIRT‐KD tumor cells had the opposite effect. These experiments indicated that SIRT1 in tumor cells could suppress anti‐tumor immunity through paracrine signaling. Additionally, we found that the expression level of SIRT1 was negatively correlated with the apoptosis rate of tumor cells (Figure [95]S2E,F, Supporting Information). These data suggested that the co‐culture system served as an excellent in vitro model for investigating the one‐way influence of tumor cells on T cells. Figure 2. Figure 2 [96]Open in a new tab SIRT1 promotes the expression of CX3CL1 in colorectal tumor cells. A,B) Elisa of effector cytokines IFNγ (A), TNFα (B) in the supernatant of activated T cells co‐culture with indicated HCT116 cells. n = 6, one‐way ANOVA test of variance, *p <0.05, **p < 0.01, ***p < 0.001. C) Activated T cells were co‐cultured with indicated HCT116 cells. Flow cytometry analysis of the ratio of PD‐1^+ cells in CD8^+ T cells. n = 7, one‐way ANOVA test of variance, **p < 0.01, ***p < 0.001. D) Activated T cells were co‐cultured with indicated HCT116 cells. Flow cytometry analysis of the ratio of Ki67^+ cells in CD8^+ T cells. n = 6, one‐way ANOVA test of variance, **p < 0.01, ***p < 0.001. E) The correlation of SIRT1 with all chemokines, analyzed by GEPIA. F) The SIRT1 and CX3CL1 levels of SIRT1‐KD and control HCT116 (left) or SIRT1‐OE and control HCT116 (right) were assessed using immunoblotting. G,H) Confocal assay of SIRT1 (green) and CX3CL1 (red) in SIRT1‐OE, SIRT1‐KD, or control HCT116, and the fluorescence intensity was calculated by Image J software. Representative images of SIRT1‐KD, SIRT1‐OE, or control HCT116 (F) and correlation matching between SIRT1 and CX3CL1 (G) are shown. Scale bars, 20µm. I) Based on the median mRNA expression value, TCGA‐COAD samples were divided into two groups: SIRT1^LowCX3CL1^Low and SIRT1^HighCX3CL1^High. Kaplan‐Meier analysis of the overall survival rate of patients in different groups. Red line: SIRT1^LowCX3CL1^Low; Blue line: SIRT1^HighCX3CL1^High. J) Multivariate logistic regression analysis of odds ratio (ORs) of different clinic‐pathological characteristics showed that compared with SIRT1^LowCX3CL1^Low profile, SIRT1^HighCX3CL1^High profile had a higher risk of large tumor grade, invasive depth, and metastasis. Next, we analyzed the correlation between SIRT1 and the expression of all chemokines using GEPIA in the TCGA database. The results demonstrated that among chemokines, CX3CL1 was the most strongly correlated with SIRT1 in CRC (Figure [97]2E). Subsequently, we performed immunoblotting and immunofluorescence analyses to confirm the positive correlation between the expression levels of SIRT1 and CX3CL1 further (Figure [98]2F–H). We then assessed whether SIRT1 and CX3CL1 could be combined to indicate CRC prognosis. CRC samples from the TCGA database were divided into high and low‐expression groups based on the median expression levels of SIRT1 and CX3CL1, respectively. The intersection of expression groups for both genes revealed that 188 samples had the same expression patterns for SIRT1 and CX3CL1 (96 samples had high expression of both SIRT1 and CX3CL1, and 92 samples had low expression of both SIRT1 and CX3CL1). Survival analysis of these samples revealed that patients with a SIRT1^HighCX3CL1^High profile had a significantly lower overall survival rate than those with a SIRT1^LowCX3CL1^Low profile (Figure [99]2I). Moreover, the upregulation of SIRT1 and CX3CL1 was significantly correlated with several aggressive clinicopathological features of CRC, including advanced tumor stage, increased lymphatic and distant metastasis rates, and increased tumor invasion depth (Figure [100]2J). These findings suggested that the SIRT1‐CX3CL1 axis was critical in promoting the progression and spread of CRC and could serve as a prognostic biomarker for identifying patients with a high likelihood of poor outcomes. 2.3. SIRT1 Activates FOXO1 to Promote CX3CL1 Expression Next, we explored the mechanisms by which SIRT1 enhances the expression of CX3CL1. First, we performed RNA sequencing (RNA‐seq) on WT and SIRT1‐OE HCT116 cells. To identify signaling pathways that SIRT1 might directly regulate, we conducted a Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the DEGs in the SIRT1‐OE group. We found significant enrichment of the FoxO signaling pathway in the SIRT1‐OE cells (Figures [101] 3A and [102]S3A, Supporting Information). The FoxO transcription factor family consists of four members: FOXO1, FOXO3, FOXO4, and FOXO6. Screening based on the cBioPortal website revealed that FOXO1 and FOXO3 exhibited the strongest correlation with SIRT1 (Figure [103]3B). GEPIA analysis indicated that the expression of CX3CL1 was significantly positively correlated with that of FOXO1 but not with that of FOXO3 in CRC patients (Figure [104]3C). Moreover, the expression of SIRT1 was positively correlated with that of FOXO1 in various gastrointestinal cancers (Figure [105]S3B, Supporting Information). To further validate the FOXO1 function, we separately performed FOXO1 or FOXO3 overexpression or knockdown experiments in HCT116 and SW480 cells and detected the expression of CX3CL1 in the culture supernatant via ELISA (Figure [106]S3C,D, Supporting Information) and western blot (Figures [107]3D,E and [108]S3F,G, Supporting Information). We observed that FOXO1 significantly affects the level of CX3CL1, while FOXO3 has a relatively minor impact on the level of CX3CL1 (Figure [109]S3E,H Supporting Information). Moreover, treatment of CRC cells with a FOXO1‐specific inhibitor (AS1842856) significantly suppressed the secretion of CX3CL1 (Figure [110]S3I, Supporting Information). These data indicated that SIRT1 promoted CX3CL1 secretion by regulating FOXO1. Figure 3. Figure 3 [111]Open in a new tab SIRT1 activates FOXO1 to promote CX3CL1 expression. A) The upregulated DEGs of SIRT1‐OE relative to SIRT1‐WT HCT116 cells were subjected to KEGG pathway enrichment analysis. The top 20 KEGG pathways with the most significant p‐values were displayed. B) Screening graph showing the relationship between SIRT1 and FOXO family members using cBioPortal in CRC samples. C) GEPIA analysis of the correlation between CX3CL1 and FOXO1 or FOXO3. D,E) FOXO1 and CX3CL1 levels of FOXO1‐KD, FOXO1‐OE, and control HCT116 (D) or SW480 (E) were assessed using immunoblotting. F,G) SIRT1 immune complexes were immunoprecipitated from HCT116 (F) or SW480 (G) cells and subjected to immunoblotting of FOXO1. H,I) Acetyl level of FOXO1 in SIRT1‐KD (H) or SIRT1 inhibitor EX527 (20 µM, 48 h) treated HCT116 cells (I) was assessed using immunoblotting. J,K) Confocal assay of SIRT1 (green) and FOXO1 (red) in SIRT1‐OE, SIRT1‐KD, or control HCT116 and the fluorescence intensity of FOXO1 in nuclear was calculated by Image J software. Representative images of SIRT1‐KD, SIRT1‐OE, or control HCT116 (J) and the statistical analysis of nuclear fluorescence intensity of CX3CL1 (K) are shown. One‐way ANOVA test of variance, ***p < 0.001. Scale bars, 20µm. L) The indicated plasmids were transfected into HCT116 cells, and the acetylation of FOXO1^WT and mutants was measured by immunoblotting. (M) CX3CL1 levels of cells in (L) were assessed using immunoblotting. Then, we used coimmunoprecipitation experiments to validate the endogenous SIRT1‐FOXO1 interaction in CRC cell lines (Figure [112]3F,G). Given that SIRT1 primarily functions as a deacetylase within cells, we treated CRC cells with shSIRT1 or the SIRT1‐specific inhibitor EX527. Both treatments resulted in elevated acetylation levels of the transcription factor FOXO1 (Figures [113]3H,I and [114]S3J,K Supporting Information). This finding indicates that SIRT1 acts as a deacetylase for FOXO1. Next, we assessed whether the acetylation level of FOXO1 affected its nuclear localization. We performed immunofluorescence staining to evaluate the subcellular localization of FOXO1 in SIRT1‐WT, SIRT1‐KD, and SIRT1‐OE HCT116 cells. The results showed that deacetylation promoted the nuclear accumulation of FOXO1, while acetylated FOXO1 exhibited significantly reduced stability within the nucleus (Figure [115]3J,K). To directly examine the impact of FOXO1 acetylation status on its function, we constructed five Flag‐tagged plasmids: FOXO1^K245R‐Flag, FOXO1^K248R‐Flag, FOXO1^K262R‐Flag, and FOXO1^K265R‐Flag and FOXO1^WT‐Flag, to simulate the deacetylated state of FOXO1. We knocked down the endogenous FOXO1 in tumor cells to minimize its impact on the experimental outcomes (Figure [116]S3L, Supporting Information). Then we transfected five plasmids and found that each of the mutated plasmids reduced the acetylation levels of FOXO1, with the arginine mutation at position 262 showing the most pronounced effect (Figure [117]3L). Subsequently, through western Blot detection, we found that the expression of CX3CL1 is negatively correlated with the acetylation levels of FOXO1 (Figure [118]3M). These results suggested that SIRT1 stabilizes FOXO1 in the nucleus via deacetylation, allowing it to exert its transcriptional effects. In summary, the above experiments demonstrated that SIRT1 deacetylates FOXO1 to promote the expression of CX3CL1 in CRC cells. 2.4. CX3CL1 Promotes Immune Suppression by Mediating the Function of Treg Cells Next, we investigated the potential role of CX3CL1 in developing an immunosuppressive microenvironment. First, GEPIA analysis revealed a positive correlation between the expression of CX3CL1 and T‐cell exhaustion markers in human CRC samples (Figure [119]S4A, Supporting Information). Then, we cocultured HCT116 cells with activated T cells in transwell chambers as previously described. We found that the exogenous addition of CX3CL1 significantly increased the expression of exhaustion‐related markers on CD8^+ T cells (Figure [120] 4A,B). Recent studies have indicated that early exhausted T cells exhibit strong proliferative capacity and retained tumor reactivity. In contrast, terminally exhausted T cells exhibit reduced proliferative capacity and decreased effector cytokines production.^[ [121]^14 ^] Correspondingly, we found that the exogenous addition of CX3CL1 reduced the proliferative capacity and suppressed the production of TNF‐α and IFN‐γ by CD8^+ T cells (Figures [122]4C and [123]S4B,C, Supporting Information). These results suggest that CX3CL1 regulates T‐cell exhaustion. Figure 4. Figure 4 [124]Open in a new tab CX3CL1 promotes immune suppression by mediating the function of Treg cells. A–C) Activated T cells co‐cultured with HCT116 cells received indicated treatment (CX3CL1, 200 ng ml^−1, 48 h), and flow cytometry analysis of the ratio of PD‐1^+ cells in CD8^+ T cells (A); the ratio of TIM‐3^+ cells in CD8^+ T cells (B); the ratio of Ki67^+ cells in CD8^+ T cells (C). n = 6, student t‐test of variance, ***p < 0.001. D) The seven‐step cancer immunity cycle scores of CX3CL1^High and CX3CL1^Low CRC samples from the TCGA database. E) Using the CICS as a phenotype, upregulated DEGs of CX3CL1High versus CX3CL1Low CRC samples were used to construct a WGCNA coexpression network; each color represents a type of module. The heatmap shows the correlation of each module with CICS. Red represents a positive correlation, while blue represents a negative correlation. *p <0.05, **p < 0.01, ***p < 0.001. F) The ClueGO plug‐in was used to perform GO enrichment for the core genes in brown modules. Each node represents a biological process; nodes with the same color have similar functions. Node size shows the significance of enrichment. G) The ClueGO plug‐in was used to perform KEGG enrichment for the core genes in brown modules. Each node represents a signaling pathway; nodes with the same color have similar functions. Node size shows the significance of enrichment. H) Activated T cells co‐cultured with HCT116 cells received indicated treatment, and flow cytometry analysis of the ratio of CD25^+CD127^− (Treg) cells and CD25^−CD127^+ (memory) cells in CD4^+ T cells. n = 6, student t‐test of variance, ***p < 0.001. I) Schematic diagram of constructing a humanized mouse subcutaneous transplant tumor model. J) NCG mice with subcutaneous xenografts (10^6 HCT116 cells) have tail vein injected CD4^+/CD8^+ mixed T cells. All mice were divided into two groups and received indicated treatment, and tumor tissues were extracted at the experimental endpoint (left). The tumor size was measured every three days using calipers to plot the tumor growth curve (right). Statistical significance was assessed by student t‐test of variance. n = 6, **p < 0.01. K–N) Tumor tissues in (J) were digested into single cells, and flow cytometry analysis of the ratio of PD‐1^+ cells in CD8^+ T cells (K); the ratio of TIM‐3^+ cells in CD8^+ T cells (L); the ratio of LAG‐3^+ cells in CD8^+ T cells (M) and the ratio of CD39^+ cells in CD8^+ T cells (N). n = 6, student t‐test of variance, ***p < 0.001. O) Tumor tissues in (J) were digested into single cells, and flow cytometry analysis of the ratio of CD25^+CD127^− (Treg) cells and CD25^−CD127^+ (memory) cells in CD4^+ T cells. n = 6, student t‐test of variance, ***p < 0.001. P) TGF‐β (left) and IL‐10 (right) levels of JMS‐17‐2 treated or control HCT116 xenografts (single cell culture supernatant after tissue lysis) were assessed by ELISA assay. n = 6, student t‐test of variance, ***p < 0.001. To investigate the mechanism by which CX3CL1 mediates T‐cell exhaustion, we calculated the cancer‐immunity cycle score (CICS) ([125]http://biocc.hrbmu.edu.cn/TIP/)^[ [126]^15 ^] using mRNA expression data for the 28 samples with the highest CX3CL1 expression and the 28 samples with the lowest CX3CL1 expression among TCGA CRC samples. The results showed that the high‐CX3CL1‐expression group had a significantly greater infiltration level of Tregs and lower T‐cell effectiveness (steps 5–7) in tumor tissue than did the low‐expression group (Figure [127]4D). These data suggested that the enhanced immunosuppressive phenotype of the CX3CL1 high‐expression group might be related to Tregs. Next, we identified the DEGs upregulated in the CX3CL1 high‐expression group compared to the low‐expression group and used the CICS as a clinical marker for weighted gene co‐expression network analysis (WGCNA). We identified 18 co‐expressed gene modules and performed a correlation analysis between these modules and the CICS phenotype. We found that most modules were related to Treg recruitment, with ten modules showing positive correlations and 5 being significantly positively correlated (Figure [128]4E). Furthermore, we conducted GO and KEGG analyses of the genes in the brown module, which was most closely associated with Treg infiltration. GO analysis revealed that these genes were enriched in processes related to immunosuppression, such as “negative regulation of leukocytes,” “negative regulation of immune effector process,” and “myeloid leukocyte‐mediated immunity” (Figure [129]4F). On the other hand, KEGG analysis revealed that these genes were enriched in classical pathways that activate Treg function, including the “JAK‐STAT signaling pathway” and “MAPK signaling pathway” (Figure [130]4G). We also found a significant positive correlation between the expression of CX3CL1 and that of the key transcription factor FOXP3, which regulates Treg function, as well as between the expression of the Treg effector molecules IL‐10 and TGFβ in human CRC tissues via GEPIA (Figure [131]S4D, Supporting Information). These results suggest that CX3CL1 helps tumor cells establish an immunosuppressive microenvironment by promoting the infiltration and enhancing the function of Tregs. We then proceeded to validate the effect of CX3CL1 on Treg cells directly via in vitro experiments. T cells isolated from PBMCs were activated with anti‐CD3, anti‐CD28 antibodies, and IL‐2. Then activated T cells were cocultured with exogenously added CX3CL1 for 48 h. The results showed that, compared to those in the control group, the proportion of Treg cells (CD4^+CD25^+CD127^−) was significantly higher, and the proportion of memory T cells (CD4^+CD25^−CD127^+) was significantly lower in the CX3CL1‐treated group (Figure [132]4H). Although CX3CL1 enhances the function of Treg cells, we could not determine whether the increase in T‐cell exhaustion induced by CX3CL1 is due to its effect or mediated by Treg cells. To address this issue, we sorted ex vivo‐activated T cells into CD4^+ T cells and CD8^+ T cells. Compared to adding CX3CL1 only in CD8^+ T cells, adding CX3CL1 in mixed CD4^+ T and CD8^+ T cells significantly increased the expression levels of exhaustion markers (Figure [133]S4E,F, Supporting Information) and attenuated proliferative capacity (Figure [134]S4G, Supporting Information) on CD8^+ T cells. We further validated that CX3CL1 promotes T‐cell exhaustion by regulating Treg function in humanized mice subcutaneously engrafted with CRC cells. Specifically, one week after the injection of HCT116 cells into the flanks of NOD/ShiLtJGpt (NCG) mice to induce subcutaneous tumor formation, in vitro activated CD8^+ T cells (1 ×  10^7 per mouse) or mixed CD4^+ T cells (5 × 10^6 per mouse) and CD8^+ T cells (5 ×  10^6 per mouse) cells from the same healthy donor were injected via the tail vein every week (Figure [135]4I). We also treated mice injected with CD8^+ T cells or mixed T cells with the CX3CL1‐specific inhibitor JMS‐17‐2 (10mg kg^−1 per day). We discovered that in mice intravenously injected with a mixture of CD4^+ T and CD8^+ T cells via the tail vein, JMS‐17‐2 significantly inhibited tumor progression (Figure [136]4J). However, the antitumor effect of JMS‐17‐2 was not pronounced in mice that were solely administered CD8^+ T cells (Figure [137]S4H, Supporting Information). At the endpoint of the in vivo experiment in which mice were injected with mixed T cells, we euthanized all the mice, extracted the tumors, and digested them into single cells for flow cytometric analysis. Firstly, we evaluated the infiltration of T cells in tumor tissues and found that JMS‐17‐2 could significantly increase the level of T cell infiltration (Figure [138]S4I, Supporting Information). The proportion of T cells was between 18%‐25%, which is essentially consistent with the abundance of T cell infiltration in surgically resected tissues from patients.^[ [139]^16 ^] Next, we found that applying JMS‐17‐2 treatment could significantly increase the proliferative capacity of CD8^+ T cells, suggesting a stronger tumor reactivity (Figure [140]S4J, Supporting Information). Notably, the administration of JMS‐17‐2 significantly alleviated CD8^+ T cell exhaustion in the mice, as evidenced by the decreased expression of exhaustion markers such as PD‐1, TIM‐3, LAG‐3, and CD39 (Figure [141]4K–N). Correspondingly, we found that JMS‐17‐2 treatment reduced Treg infiltration and a significant increase in the proportion of memory T cells (Figure [142]4O). We also conducted ELISA tests on the primary cell supernatant and found that IL‐10 and TGFβ levels were significantly decreased in the JMS17‐2‐treated group, indicating diminished Treg function (Figure [143]4P). To further validate the immunomodulatory effects of CX3CL1‐CX3CR1 signaling blockade, we administered neutralizing antibodies targeting CX3CL1 (Quetmolimab, 200 µg per mouse/3 days) to assess the immunosuppressive effects of CX3CL1‐CX3CR1 signal blocking in humanized mouse models injected with a mixture of CD4^+ T and CD8^+ T cells. We observed that, compared to the control group, Quetmolimab treatment exhibited a significant anti‐tumor effect (Figure [144]S5A, Supporting Information). The immune microenvironment analysis revealed that Quetmolimab treatment significantly reduced the expression of exhaustion markers (Figure [145]S5B,C, Supporting Information) and increased the tumor reactivity of CD8^+ T cells (with elevated expression of Ki67, TNF, and IFN) (Figure [146]S5D–F, Supporting Information), indicating a reduction in CD8^+ T cell exhaustion. Correspondingly, the blockade of CX3CL1 led to a decrease in the infiltration of Treg cells (Figure [147]S5G, Supporting Information). Taken together, these findings indicated that CX3CL1 enhanced the function of Treg cells to suppress anti‐tumor immunity. 2.5. CX3CL1 Enhances the Function of Treg Cells by Promoting the TNFRSF9 Phenotype To further investigate the role of CX3CL1 signaling in Treg cells, we first examined the expression of CX3CL1 and its unique receptor CX3CR1 in different Treg populations. We examined scRNA‐seq data ([148]GSE108989) from tissues of 12 CRC patients and found that Treg cells from PBMCs and adjacent normal tissue hardly expressed CX3CR1. In contrast, a subset of Treg cells infiltrating the tumor tissue exhibited high expression of CX3CR1 (Figure [149]S6A, Supporting Information). Next, we integrated all pairs of Treg cell samples. The UMAP analysis revealed distinct origins of Treg cell clusters: Cells in Clusters 0 and 3 were primarily from PBMCs and adjacent normal tissues, making up 31.67% and 38.47% of these subgroups, respectively, with only 10.8% from CRC tissues. In contrast, Clusters 1 and 4 were mainly tumor tissue‐infiltrating Treg cells, with 29.72% from CRC, while contributions from adjacent tissues and PBMCs were minimal at 3.48% and 5.47%, respectively (Figures [150] 5A and [151]S6B, Supporting Information). The above results indicated that the phenotype of tumor‐infiltrating Treg cells significantly differed from that of Tregs derived from PBMCs or adjacent normal tissues. Figure 5. Figure 5 [152]Open in a new tab CX3CL1 enhances the function of Treg cells by promoting the TNFRSF9 phenotype. A) UMAP plot based on the scRNA‐seq data ([153]GSE108989) of Treg cells sourced from peripheral blood, adjacent normal tissue, and cancer tissue. Create visual representations that reflect the origin of the samples. Determine and quantify the percentages of cells within Clusters 0, 3 (encircled by the purple dotted line) and Cluster 1, 4 (encircled by the blue dotted line) for each sample category in relation to the overall number of cells. NTR, Tregs from adjacent normal colorectal tissues; PTR, Tregs from peripheral blood; TTR, Tregs from CRC. B–D) Feature plots of indicated genes across all Treg subclusters in (A). E) scRNA‐seq of CD45^+ immune cells extracted from resected tumor tissues of three primary‐care CRC patients. UMAP plot of Tregs (CD4^+FOXP3^+, n = 4628), identifying nine distinct subgroups. Each subgroup was named by marker genes calculated using the Seurat package. F) Feature plots of indicated genes across all Treg subclusters in (E). G) UMAP and extrapolated future state of cells (overlaid arrows) based on RNA velocity. H) Putative driver genes of TNFRSF9^+ Treg are identified by high likelihoods. Phase portraits (left) and expression dynamics (right, green represents upregulation, while red represents downregulation) for these driver genes characterize their activity. I) Heatmap shows the GEPIA analysis of the correlation between putative driver genes in (H) and all chemokine receptors. J) The expression level of putative transcription factors in CX3CL1 treated or control CD4^+ T cells was assessed using immunoblotting. K) The expression level of BTG2 and SATB1 in CX3CL1 inhibitor JMS‐17‐2 treated or control CD4^+ T cells was assessed using immunoblotting. L,M) Co‐culturing CD4^+ T cells from different groups, as illustrated, with HCT116 cells. The ratio of TNFRSF9^+ cells in CD25^+ T cells was analyzed by flow cytometry. n = 6, student t‐test of variance, **p < 0.01, ***p < 0.001. (N) Exogenous addition of CX3CL1 significantly increased the IL‐10 (left) and TGF‐β (right) levels of CD4^+ T cells co‐cultured with HCT116. n = 6, student t‐test of variance, ***p < 0.001. A previous study reported that TCF‐1‐deficient Treg cells strongly suppressed T‐cell proliferation and cytotoxicity.^[ [154]^11 ^] Correspondingly, Cluster 0 and Cluster 3 Treg cells exhibited high TCF7 expression (encoding the TCF1), while Cluster 1 and Cluster 4 Treg cells barely expressed TCF7 (Figure [155]5B). Moreover, we found that CX3CR1 was primarily expressed in Treg cells from Cluster 4 (Figure [156]5C). Notably, TNFRSF9^+ Tregs are a subset that has been demonstrated to play a crucial immunosuppressive role in various tumors.^[ [157]^10 ^] Our analysis showed that Treg cells in both Cluster 1 and Cluster 4 were in the TNFRSF9^+ Treg subset (Figure [158]5D). Because of the overlap between CX3CR1^+ cells and TNFRSF9^+ cells, these results suggested that the CX3CL1‐CX3CR1 signaling pathway might be involved in shaping the TNFRSF9^+ Treg phenotype. Lipid synthesis and metabolism are closely associated with Treg differentiation and functional enhancement.^[ [159]^17 ^] We identified DEGs between Treg cells in Cluster 4 and those in Clusters 0 and 3, and subsequently conducted GSEA. The results indicated that lipid biosynthetic and lipid metabolism processes are more active in Tregs from Cluster 4 (Figure [160]S6C, Supporting Information). Moreover, genes associated with the “receptor signaling pathway via JAK‐STAT” pathway were significantly enriched in Cluster 4 (Figure [161]S6D, Supporting Information). The above data suggested that CX3CL1‐CX3CR1 signaling might promote Treg function. To explore the impact of CX3CL1‐CX3CR1 signaling on the phenotype of Tregs, we performed scRNA‐seq on CD45^+ immune cells sourced from three resected CRC samples. We then integrated and clustered the CD4^+FOXP3^+ cells (n = 4628) and identified nine subgroups after batch effect removal (Figure [162]5E). Consistent with the above results, we observed similar expression patterns of CX3CR1 and TNFRSF9, and the CX3CR1^+ cells did not express TCF7 (Figure [163]5F). Subsequent RNA velocity analysis of the scRNA‐seq data indicated that TCF7^+ Treg cells served as the starting point for the differentiation of various phenotypes of Treg cells in the tumor microenvironment. TNFRSF9^+ Treg cells were identified as a terminal differentiation phenotype (Figure [164]5G). Next, by individual gene dynamics screening of transcription factors, we identified five potential transcription factors that correlated most strongly with the differentiation of TCF7^+ Treg cells to TNFRSF9^+ Treg cells: NFKBIA, BTG2, JUND, SATB1, and FOSB (Figure [165]5H). To further clarify the transcription factors associated with the CX3CL1‐CX3CR1 signaling pathway, we calculated the correlation of these transcription factors with the expression of all chemokine receptors in CRC samples from the TCGA database. The results indicated that CX3CR1 strongly correlates with BTG2 and SATB1 (Figure [166]5I). Next, we activated T cells isolated from PBMCs and cocultured them with HCT116 cells after sorting the CD4^+ T cells. We observed that the exogenous addition of CX3CL1 increased the expression of SATB1 and BTG2 in CD4^+ T cells, while its impact on the other three transcription factors was minimal (Figures [167]5J and [168]S6E, Supporting Information). Simultaneous JMS17‐2 treatment reduced the expression of SATB1 and BTG2 (Figures [169]5K and [170]S6F, Supporting Information). To further validate the promoting effect of BTG2 and SATB1 on the TNFRSF9^+ Treg phenotype, we constructed lentiviruses for overexpressing these two transcription factors. T cells activated by anti‐CD3, anti‐CD28, and IL‐2 were transfected with empty vector, BTG2‐OE, SATB1‐OE, and a combination (BTG2‐OE+SATB1‐OE) lentivirus. The overexpression of transcription factors was verified by western blotting (Figure [171]S6G, Supporting Information). After sorting CD4^+T cells by flow cytometry, we co‐cultured them with tumor cells according to the illustrated treatment. We then detected the proportion of TNFRSF9^+ Treg cells after 72 h. The results indicated that overexpressing BTG2 and SATB1 significantly increased the differentiation of the TNFRSF9^+ Treg phenotype, and the combined overexpression achieved an effect comparable to that of CX3CL1 stimulation (Figures [172]5L,M and [173]S6H,I, Supporting Information). These results suggested that BTG2 and SATB1 are downstream effector molecules of the CX3CL1‐CX3CR1 signaling pathway. Finally, we co‐cultured CD4^+ T cells with tumor cells for 48 h and found that the exogenous addition of CX3CL1 significantly promoted the secretion of IL10 and TGFβ (Figures [174]5N and [175]S6J, Supporting Information). Therefore, these results indicated that the CX3CL1‐CX3CR1 signaling pathway facilitates the induction of the TNFRSF9^+ Treg phenotype by activating BTG2 and SATB1. 2.6. The Clinical Relevance of CX3CL1 Signaling To explore the potential of CX3CL1 as a prognostic indicator in clinical samples and validate its correlation with Treg infiltration, we conducted multiplex immunofluorescence (mIHC) staining of a tissue microarray containing 100 CRC tissue samples with antibodies against CX3CL1, CD4, CD25, and PANCK (a tumor cell marker). Given that CD25^+ Tregs are an independent risk factor for a poor prognosis in patients with CRC, we used CD4 and CD25 as Treg markers instead of CD4 and FOXP3.^[ [176]^18 ^] By the 7th edition of the American Joint Committee on Cancer (AJCC) cancer staging system, we categorized tumor tissues with a grade