Abstract Ulcerative Colitis (UC) is an inflammatory disorder characterized by chronic intestinal inflammation and immune dysregulation. Despite a clear association between cellular senescence and chronic inflammation and immune dysregulation, the mechanisms underlying cellular senescence in UC remain unclear. We screened differentially expressed genes (DEGs) associated with cellular senescence in multiple UC datasets, performed immune infiltration analysis, and constructed clinical diagnostic models. Additionally, we investigated the relationship between key genes related to cellular senescence and disease remission in UC patients undergoing biologic therapy, validating their expression in a single-cell dataset. We identified six DEGs associated with cellular senescence (TWIST1, IGFBP5, MME, IFNG, ME1, FOS). Immune infiltration results indicated strong correlations of four of these genes with immune cells and pathways. Through WGCNA, GO, and KEGG analyses, we found that gene modules strongly associated with the expression of hub genes in cellular senescence were enriched in inflammation-related pathways. In the single-cell dataset, the expression of these six key genes exhibited similarities with Immune infiltration results. Additionally, we constructed a nomogram using these six genes for diagnosing UC, demonstrating good diagnostic capability and clinical efficacy. Kaplan–Meier survival analysis revealed a significant association between changes in the expression levels of these cell genes and disease remission in UC patients undergoing biologic therapy. This study utilizes bioinformatic analysis and machine learning to identify and analyze features associated with cellular senescence in multiple UC datasets. It provides insights into the role of cellular senescence in the premature onset of intestinal aging in UC and offers new perspectives for exploring novel therapeutic targets. Keywords: Ulcerative colitis, Cellular senescence, GEO dataset, Biologics, Immune, Inflammatory bowel disease Subject terms: Immunology, Gastrointestinal diseases, Inflammatory bowel disease, Ulcerative colitis Introduction Ulcerative Colitis (UC) is a predominant inflammatory bowel disease (IBD) affecting the colon and rectum, characterized by persistent and recurrent chronic intestinal inflammation with symptoms of mucous bloody stools^[38]1. The clinical course of UC is unpredictable, influenced by various factors such as genetic inheritance, dietary habits, environmental factors, immune system dysregulation, and gut microbiota imbalance^[39]2. In recent years, the global incidence of UC has been steadily increasing, posing a serious threat to public health, especially in developing countries^[40]2,[41]3. Currently, UC has no cure, relying mainly on long-term use of immunosuppressive drugs or steroids to control disease activity, which often entails significant side effects^[42]4. Thus, in-depth investigation into the pathogenesis of UC is essential for discovering new therapeutic targets and diagnostic markers, significantly impacting the alleviation of clinical symptoms in UC patients and improving the efficacy of UC drug treatments. UC is an immune system dysregulation-related inflammatory disease of the intestines. Disease progression in UC is closely associated with cellular senescence, although their interplay remains largely unexplored^[43]5. Cellular senescence is a cellular state linked to various physiological processes and a broad spectrum of age-related diseases. It is triggered by diverse stress signals throughout the cell’s lifespan, characterized by cell cycle arrest, a senescence-associated secretory phenotype (SASP), metabolic imbalance, and macromolecular damage^[44]6,[45]7. In a healthy immune system, aging cells within the intestines are effectively cleared to maintain gut homeostasis^[46]8. However, in UC, disrupted immune system homeostasis leads to the accumulation of senescent cells, which promote an inflammatory state associated with aging and exacerbate intestinal inflammation^[47]9,[48]10. Chronic inflammation in the intestines results in the continuous production of inflammatory factors, depleting adaptive immune responses, eventually leading to immune senescence. Immune senescence and intestinal inflammation can operate simultaneously, forming a detrimental feedback loop^[49]11. Additionally, aging epithelial cells weaken the intestinal mucosal barrier, allowing bacterial translocation, further worsening local and systemic inflammation^[50]12,[51]13. Although aging is commonly considered a major issue for the elderly population, patients with UC experiencing recurrent intestinal inflammation and immune system dysregulation may accelerate the onset of intestinal aging. Therefore, evaluating the involvement of genes related to cellular senescence in UC intestinal inflammation and immunity helps elucidate the mechanistic role of cellular senescence in UC progression. In this study, we downloaded three datasets containing gene expression data from UC colonic mucosa ([52]GSE107499, [53]GSE87466, [54]GSE59071) from the GEO database. After merging and screening for differentially expressed genes (log|FC|> 1, p-value < 0.05), we intersected these genes with those related to cellular senescence, obtaining 10 differentially expressed genes (all upregulated). Subsequently, we utilized lasso regression to further narrow down the genes obtained in the previous step, ultimately identifying six upregulated genes related to cellular senescence (Fig. [55]1). Additionally, we observed the immune infiltration landscape in the UC intestines and constructed a nomogram with these six key genes for diagnosing UC, assessing the clinical efficacy of the diagnostic model. Finally, we downloaded dataset [56]GSE73661 to validate the performance of these six genes in UC biologic therapy (vedolizumab (VDZ) and infliximab (IFX)), and further validated the expression changes of these six genes in UC single-cell RNA dataset ([57]GSE221987). Fig. 1. [58]Fig. 1 [59]Open in a new tab The flow chart of the study, including experimental grouping design and research process. UC Ulcerative colitis, CS Cell senescence. Materials and methods Data source and clinical validation experiments of key genes The microarray data used for our analysis was obtained from the Gene Expression Omnibus (GEO) database ([60]https://www.ncbi.nlm.nih.gov/geo/), with the accession numbers [61]GSE107499, [62]GSE87466, [63]GSE59071, [64]GSE73661, and [65]GSE221987. A total of 312 samples of intestinal mucosal tissue were selected, including 236 samples from UC active patients and 76 samples of normal intestinal mucosal tissue. The gene expression data was normalized and merged for subsequent analysis. The [66]GSE73661 dataset was chosen to evaluate the impact of vedolizumab and infliximab on the expression of hub genes, identifying the utility of hub genes in predicting disease remission after biologic therapy. The [67]GSE221987 dataset, which includes single-cell RNA sequencing results of colonic mucosa from four UC patients, was used to validate the expression patterns of key genes across various cells. Clinical specimens from UC patients undergoing colonoscopy were collected at the Second Xiangya Hospital of Central South University for experimental detection. This procedure was approved by the Ethics Committee of the Second Xiangya Hospital of Central South University (Approval No: 2022-155). We confirm that all methods were carried out in accordance with relevant guidelines and regulations. Informed consent was obtained from all subjects. We collected intestinal mucosal tissues from 10 UC patients in the active phase, with basic information shown in Supplementary Table [68]1. Normal tissues were taken from the normal colonic mucosa of the same UC patients. At week 8 after patients received regular vedolizumab monoclonal antibody treatment, we performed colonoscopy on these UC patients to assess the presence of drug remission. Response was defined as endoscopic mucosal healing (Mayo endoscopic subscore 0 or 1) and was assessed for vedolizumab at W8. The qRT-PCR primer sequences are shown in Supplementary Table 2. Acquirement of differentially expressed cell senescence related genes We downloaded 432 and 949 genes associated with cellular senescence from the MSigDB (Molecular Signatures Database) genetic database^[69]14 and the CellAge genetic database^[70]15 , respectively. After taking the intersection of the two sets, we obtained a total of 134 genes related to cellular senescence for subsequent analysis. Using the criteria of fold change (log|FC|) > 1 and p < 0.05 for differential gene expression, we applied the “limma” R package to identify genes with differential expression in the intestinal mucosa of UC patients. We then compared the differentially expressed genes with the cell senescence related genes and found a total of 10 upregulated genes and we created an Upset diagram to illustrate the results. Diagnostic model built based on Lasso regression The ten upregulated genes identified previously underwent Lasso regression for further screening. Utilizing a tenfold cross-validation method, we iteratively developed a high-performance model with minimal variables. From these, six genes were selected and used to construct a diagnostic model (Nomogram). The expression matrix was randomly divided into training and validation sets in a 7:3 ratio. The training set was employed for model establishment, while the validation set assessed model performance. Validation of the cell senescence related genes signature A predictive model was developed using six hub genes associated with cell senescence to distinguish UC patients from healthy individuals. The “pROC” package in R software was utilized to generate a receiver operating characteristic (ROC) curve and compute the area under the curve (AUC), which validated the performance of the predictive feature. Calibration curves were plotted to evaluate the calibration of the nomogram prediction model. Decision curve analysis (DCA) and clinical impact curve were performed to determine the clinical usefulness of the nomogram. DCA compared the net benefits of each prediction model at different threshold probabilities. Net benefit calculations considered the trade-off between missed interventions and the adverse effects of unnecessary interventions. Analyses of immune landscape and immune correlation Single-sample gene set enrichment analysis (ssGSEA) is a machine learning algorithm used to analyze the enrichment of specific pathways in gene expression within individual samples^[71]16. The “CIBERSORT” algorithm is a gene expression-based deconvolution method that determines the cellular composition of tissues using the LM22 reference set^[72]17. We performed separate “CIBERSORT” and “ssGSEA” analyses on the selected 6 cell senescence related differentially expressed genes. We assessed the correlation between these genes and immune cell type enrichment scores, considering a p-value < 0.05 as statistically significant. Based on the criteria of having a correlation score |(Ps)|≥ 0.60 in CIBERSORT results and ssGSEA results, we identified 4 cell senescence related genes that exhibited a statistically significant association with immune functions, and these 4 genes were used for subsequent WGCNA analysis. WGCNA analysis WGCNA (Weighted Gene Co-expression Network Analysis) was applied to construct a gene co-expression network based on four genes identified through immune infiltration^[73]18. The module Preservation function within WGCNA assessed the preservation of network modules across cohorts and evaluated the influence of missing values. Z summary composite preservation scores were computed using 200 permutations, comparing each cohort or missing value threshold to the consensus network^[74]19. Functional annotation and pathway enrichment analysis Gene Ontology (GO) enrichment analysis was performed to annotate gene sets into the biological process (BP), molecular function (MF), and cellular component (CC) terms^[75]20. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was conducted to identify the signaling pathways related to DEGs. p < 0.05 was set as the criterion to identify the significantly enriched GO terms and KEGG pathways^[76]20. Single-cell RNA-seq (scRNA) analysis Single-cell data analysis was conducted using the ‘Seurat’ R package. After importing the data, standardization was performed, filtering out cells with UMI counts exceeding 2500 or below 200, as well as cells with > 5% mitochondrial content^[77]21. From each sample, 2000 highly variable genes were selected for downstream analysis. The FeatureScatter function was employed to observe the correlation between different variables. Following clustering, 12 cell clusters were obtained, and manual cell annotation was performed. Figures illustrating the clusters, as well as marker expression in the low-dimensional space (UMAP), were generated using Seurat visualization functions^[78]22. Code examples for scRNA-seq can be found in the supplementary files. Statistical analysis R software (version 4.2.0) was used for data processing and statistical analysis. Expression differences between patients with UC and controls were assessed using the Wilcoxon signed-rank and Kruskal–Wallis tests. Pearson and Spearman correlation analyses were employed to assess the statistical correlation between parametric and non-parametric variables. The p-value < 0.05 was considered statistically significant. (*p < 0.05, **p < 0.01, ***p < 0.001). Results Identification of cell senescence related DEGs Firstly, we merged three UC datasets and removed batch effects (Fig. [79]2A,B). The PCA plot demonstrates the intra-group clustering of UC and control groups (Fig. [80]2C). Filtering under the criteria of log|FC|> 1 and p < 0.05, we identified 489 upregulated genes and 220 downregulated genes (Fig. [81]2D). The heatmap illustrates the differential expression of all genes between the two groups (Fig. [82]2E). Next, we downloaded genes associated with cellular senescence from the MSigDB and CellAge databases, resulting in 134 cellular senescence genes (Fig. [83]3A). Intersection of these genes with 709 differentially expressed genes in UC yielded 10 differentially expressed genes, all upregulated (Fig. [84]3A). To further refine our target genes, we subjected the expression data of these 10 genes to Lasso regression. After selecting a model with excellent performance and the minimum number of variables, we identified six key genes (Fig. [85]3B,C). The heatmap depicts the upregulation of these six genes in UC (Fig. [86]3D). Correlation analysis reveals internal associations among these six differentially expressed genes (Fig. [87]3E), and the box plot illustrates the expression level variations of target genes between UC and normal control groups (Fig. [88]3F). Fig. 2. [89]Fig. 2 [90]Open in a new tab The three datasets [91]GSE107499, [92]GSE87466, and [93]GSE59071 were combined and normalized to create a new dataset. (A) Box plots before merging. (B) Box plots after merging. (C) Principal component analysis of the new dataset. (D) Volcano plot of differentially expressed genes (DEGs), where the red nodes represent the 489 significantly upregulated DEGs, and the blue nodes show the 220 downregulated DEGs based on log2 |FC|> 1 and p < 0.05. (E) Heatmap illustrating the expression patterns of all DEGs in individual Ulcerative Colitis (UC) and normal samples. DEGs Differentially expressed genes. Fig. 3. [94]Fig. 3 [95]Open in a new tab Selection of differentially expressed genes related to cellular senescence. (A) UpSet plot showing the intersection quantity of upregulated and downregulated genes with cellular senescence genes, respectively. (B) LASSO coefficient distribution plot of differentially expressed genes related to cellular senescence. (C) Cross-validation plot of penalty terms, indicating the retention of 7 variables when the error is minimized; corresponding to the position of the left dashed line. To avoid overfitting and simplify the model, the selected standard error does not exceed 1, retaining 6 variables, corresponding to the position of the right dashed line. (D) Heatmap displaying the expression of these 6 key genes in the merged dataset. (E) Correlation heatmap of these 6 key genes. (F) Boxplot showing the expression level variation of target genes between UC and normal control groups. The immune infiltration landscape of UC and the selection of immune-related cell senescence DEGs Cellular senescence is associated with immune dysregulation, but the connection between changes in the immune microenvironment in the UC intestinal tract and cellular senescence remains unclear. Therefore, to further understand the pathogenesis of UC, we utilized two different algorithms, CIBERSORT and ssGSEA, to compare the differences in immune cell infiltration levels in the intestinal mucosa of UC patients. CIBERSORT analysis revealed that both UC and the normal control group showed activation of T cells in the intestinal tract. However, the infiltration level of NK cells in UC patients was significantly lower than in the control group. Additionally, there was a significant increase in M1-type pro-inflammatory macrophages and neutrophil infiltration in UC (Fig. [96]4C). In ssGSEA results, the infiltration level of CD8 T cells in UC patients was significantly higher than in the control group (Fig. [97]4D), which is also a hallmark of cellular senescence, indicating the presence of excessive cellular senescence in the UC intestinal tract. To further elucidate the connection between cellular senescence and the occurrence and development of UC, we conducted correlation analysis between the six key genes selected in the previous step and the abundance of immune cells. As expected, these six key genes showed a strong correlation with immune cells (Fig. [98]4A,B). Based on the correlation coefficients and p-values from the results of the two different algorithms, we selected four genes (TWIST1, IFNG, IGFBP5, MME) with the strongest correlations for WGCNA to construct a co-expression network. Fig. 4. [99]Fig. 4 [100]Open in a new tab Immune infiltration landscape of UC. (A) Correlation matrix of enrichment scores for cellular senescence-related DEGs calculated by CIBERSORT. (B) Correlation matrix of enrichment scores for cellular senescence-related DEGs calculated by ssGSEA. (C) Comparison of immune cell infiltration in UC and normal control groups calculated by CIBERSORT. (D) Comparison of immune cell infiltration in UC and normal control groups calculated by ssGSEA. (*p < 0.05, **p < 0.01, ***p < 0.001). Construction of weighted gene co-expression network and functional annotation of key modules for immune-related cell senescence DEGs Due to the association between cellular senescence and UC, it is essential to utilize WGCNA to identify genes co-expressed with target genes. We selected a soft-threshold of 8 (based on the scale-free topology criterion with R2 = 0.80) to construct a scale-free network. The adjacency matrix was then transformed into a TOM (Topological Overlap Matrix) matrix (Fig. [101]5A), which represents the similarity between nodes by considering weighted correlations. A total of 26 gene modules were obtained (Fig. [102]5B), with all modules having Z-scores greater than 10 (Fig. [103]5C). Correlation analysis was performed between all modules and hub genes, revealing strong correlations between the turquoise, red, pink, blue and brown modules with these 4 genes (Fig. [104]6A). Subsequently, differential gene analysis was conducted on the genes belonging to these five modules, using a threshold of |(FC)|> 1 and p < 0.05 (Fig. [105]6C). This analysis resulted in the identification of 662 differentially expressed genes, including 435 upregulated genes and 227 downregulated genes (Fig. [106]6B). Finally, "GO and KEGG enrichment analysis" was conducted to explore the potential functions and mechanisms associated with UC using these DEGs. The majority of biological processes were found to be enriched in leukocyte migration, and the GO molecular function analysis revealed enrichment in cytokine activity (Fig. [107]6D). KEGG enrichment analysis of these DEGs demonstrated significant enrichment in inflammation-related pathways (Fig. [108]6E), aligning with the known pathogenesis of UC. Fig. 5. [109]Fig. 5 [110]Open in a new tab WGCNA analysis. (A) Relationship between the scale-free fitting index and various soft-thresholding powers, as well as the relationship between average connectivity and various soft-thresholding powers. (B) Hierarchical clustering dendrogram of genes, with different colors representing different modules. (C) Module preservation calculated using a composite Zscore. The blue dashed line indicates a Zscore of 2, and the dark green dashed line indicates a Zscore of 10. Modules preserved above this threshold are considered to have high statistical significance. Fig. 6. [111]Fig. 6 [112]Open in a new tab Four immune-related cellular senescence-associated differentially expressed genes (TWIST1, IGFBP5, MME, IFNG). (A) Pearson correlation analysis results between merged modules and immune-related cellular senescence DEGs. (B) Differential analysis of gene modules (5 in total) with Ps > 0.6, visualized using a heatmap (|log2 FC|> 1, p < 0.05). (C) Volcano plot showing the expression of differentially expressed genes. (D) Gene Ontology (GO) analysis. (E) Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis. Constructing and validating a nomogram for the diagnosis of UC We employed ROC analysis to assess the ability of these six key genes to distinguish between UC patients and healthy individuals. The results demonstrated good predictive value for diagnosing UC, with AUC values exceeding 0.75 for each gene (Fig. [113]7A,B). To further enhance the diagnostic capability, we randomly divided the expression data of these six genes into training and validation sets at a 7:3 ratio. In the training set, we constructed a nomogram for diagnosing UC (Fig. [114]7C). Calibration curves of the generated model showed minimal deviation from the ideal line, indicating good diagnostic consistency and high calibration accuracy (Fig. [115]7D). The c-indices for the nomogram in the training and validation sets were 0.9551 and 0.9973, respectively (Fig. [116]8A,B). Decision Curve Analysis (DCA) was used to evaluate the clinical utility of the model by assessing the net benefit of interventions based on model predictions. DCA indicated that, as the threshold probability varied, the net benefit of intervention based on model predictions exceeded that of complete intervention or non-intervention, suggesting greater net benefit from clinical interventions guided by this decision curve (Fig. [117]8C). Clinical impact curves were generated to analyze the number of patients classified as high-risk and the true positive patients at various risk thresholds (Fig. [118]8D). To strike a balance between high net benefit and a low false-positive rate, DCA was combined with clinical impact curves. Calibration suggested that setting the risk threshold for UC at 0.70 or higher could provide higher clinical benefits and a lower false-positive rate for the entire population under consideration. Fig. 7. [119]Fig. 7 [120]Open in a new tab Construction of the nomogram for diagnosing UC. (A) ROC analysis demonstrating the diagnostic capability of the six key genes for UC. (B) AUC values. (C) Nomogram construction, with red dots representing randomly selected samples for demonstration. (D) Model calibration curve indicating a small deviation between the line and the calibrated line from the ideal line, indicating good diagnostic consistency and high calibration accuracy. Fig. 8. [121]Fig. 8 [122]Open in a new tab Validation of the nomogram for diagnosing UC. (A) Diagnostic capability of the model for UC in the training set (AUC = 0.9551). (B) Diagnostic capability of the model for UC in the test set (AUC = 0.9973). (C) Decision curve analysis of the nomogram prediction model, with the light red curve representing the 95% confidence interval; the net benefit of intervention based on the model’s predicted values is higher than full intervention or no intervention as the threshold probability changes. (D) Clinical impact curve of the nomogram prediction model, where the red solid line represents the number of UC patients classified by the nomogram at each risk threshold among every 1000 patients, and the blue dashed line represents the actual number of UC patients at each risk threshold. Differential expression and predictive value of 6 cell senescence genes in the intestinal mucosa of UC patients before and after biologic therapy The [123]GSE73661 dataset comprises gene expression data from the intestinal mucosa of UC patients before and after treatment with vedolizumab (VDZ) and infliximab (IFX). The dataset includes 12 individuals in the healthy control group, 23 individuals receiving IFX treatment, and 37 individuals receiving VDZ treatment. In UC patients undergoing IFX treatment, ROC analysis revealed a good predictive ability of the six genes for the effectiveness of biologic therapy. Except for FOS, the AUC values for the other genes exceeded 0.70 (Fig. [124]9A,B). Consistent with previous results, the expression of these six genes was upregulated in these UC patients (Fig. [125]9C–H). In non-responsive patients after IFX treatment, the expression of these six genes showed no significant change compared to pre-treatment levels. However, in patients responding to IFX treatment, the expression of the other five genes, except for FOS, was downregulated compared to pre-treatment levels, with no significant difference from the expression levels in the normal control group. Similarly, in UC patients receiving VDZ treatment, ROC analysis showed a good predictive ability of the six genes for the effectiveness of biologic therapy, with AUC values exceeding 0.70 for all genes except FOS (Fig. [126]10A,B). Again, these six genes were upregulated in these UC patients (Fig. [127]10C–H). In non-responsive patients after VDZ treatment, the expression of these six genes showed no statistically significant change compared to pre-treatment levels. However, in patients responding to VDZ treatment, the expression of the other five genes, except for FOS, was downregulated compared to pre-treatment levels, but only MME and TWIST1 expression levels dropped to normal levels. Due to limited data and the lack of specific follow-up time data for patients undergoing IFX treatment, we only used data from patients undergoing VDZ treatment for survival analysis. In the survival analysis, outcomes were defined as disease remission, with gene expression in the post-treatment matrix classified as ‘Yes’ if it was below the average pre-treatment expression level; otherwise, it was classified as 'No.' KM analysis showed that, except for FOS, the expression decrease of the other five genes was time-dependent with disease remission after VDZ treatment (Fig. [128]11A–F). Fig. 9. [129]Fig. 9 [130]Open in a new tab Expression changes of these 6 key genes in UC patients before and after IFX treatment. (A) ROC analysis showing the diagnostic capability of these genes for predicting treatment effectiveness. (B) AUC value. (C–H) Changes in the expression of these genes before and after treatment. Fig. 10. [131]Fig. 10 [132]Open in a new tab Expression changes of these 6 key genes in UC patients before and after VDZ treatment. (A) ROC analysis showing the diagnostic capability of these genes for predicting treatment effectiveness. (B) AUC value. (C–H) Changes in the expression of these genes before and after treatment. Fig. 11. [133]Fig. 11 [134]Open in a new tab Kaplan–Meier analysis depicting the probability of an effective response to VDZ treatment over time based on the expression changes of the 6 central genes. In the survival analysis, outcomes were defined as disease remission. Genes with post-treatment expression levels below the pre-treatment average were classified as "Yes," otherwise as "No." The shaded area in the graph represents the 95% confidence interval. To further explore the indicative role of cellular senescence in VDZ treatment, we used the ‘ConsensusClusterPlus’ package to cluster patients undergoing VDZ treatment based on the expression of key genes. The results showed that patients could be clustered into two groups (Fig. [135]12A–E), identified as the high-risk and low-risk groups. KM-plot results suggested that over time, patients in the high-risk group had a significantly lower response rate to VDZ biologic therapy compared to the low-risk group (Fig. [136]12F). Fig. 12. [137]Fig. 12 [138]Open in a new tab Consensus cluster analysis results. (A) Cluster matrix heatmap (K = 2). (B) Cluster matrix heatmap (K = 9). (C) Delta Area Plot. (D) Cumulative Distribution Function (CDF) plot, showing optimal clustering at K = 2. (E) Tracking Plot. (F) Stratification of all patients into high-risk and low-risk groups, with the high-risk group demonstrating significantly lower responsiveness to VDZ biological therapy compared to the low-risk group. Additionally, we detected the expression of these six key genes in the colonic mucosa of 10 UC patients treated with VDZ. Consistent with the bioinformatics analysis results, the expression of these six genes was significantly elevated in the inflammatory mucosa of UC patients (Supplementary Fig. [139]1). In the colonic mucosa of the six UC patients who achieved endoscopic remission after treatment, the expression of TWIST1, IGFBP5, MME, and ME1 significantly decreased, while the expression of FOS and IFNG did not show a significant decrease. In the colonic mucosa of the four UC patients who did not show a noticeable response to treatment, the expression of these six genes did not change significantly compared to before treatment. The expression of 6 cell senescence genes in scRNA analysis We downloaded a single-cell RNA dataset containing four UC patients ([140]GSE221987) and performed single-cell data analysis using the ‘Seurat’ R package. The quality control plot displayed gene expression in each cell, including mitochondrial gene expression (Fig. [141]13A). The FeatureScatter function was employed to observe correlations between different variables (Fig. [142]13B). After clustering, 12 cell clusters were obtained, and manual cell annotation was performed (Fig. [143]13C). Similar to the previous results on immune infiltration, the expression of these six genes showed significant differences across various immune cell types. Additionally, TWIST1 and IGFBP5 were highly expressed in stem cells (Fig. [144]14A,C), MME showed high expression in neutrophils (Fig. [145]14E), IFNG exhibited high expression in CD4 T cells (Fig. [146]14B), ME1 was highly expressed in epithelial cells and macrophages (Fig. [147]14D), and FOS was expressed in various cell types (Fig. [148]14F). Fig. 13. [149]Fig. 13 [150]Open in a new tab Single-cell RNA analysis results. (A) Quality control plot (QC). (B) Feature Scatter plot displaying the correlation between different variables. (C) Cell annotation plot. Fig. 14. [151]Fig. 14 [152]Open in a new tab Validation of the expression of 6 cellular senescence genes in the single-cell dataset, consistent with immune infiltration results. Discussion UC is a chronic inflammatory bowel disease characterized by recurrent inflammation primarily associated with immune dysregulation in the intestinal tract^[153]23. Recently, the association between cellular senescence and immune-related inflammation has gained clarity. Senescent cells, which cease division but secrete damaged and metabolically altered molecules, often resist apoptosis^[154]24. These cells frequently release cytokines via paracrine signaling, impacting tissue remodeling, promoting tumorigenesis, and sustaining chronic inflammation^[155]25. Identifying senescent cells molecularly presents challenges, as the senescent state induced by various triggers exhibits heterogeneity across tissues^[156]26. Therefore, understanding the role of cellular senescence in UC development is pivotal. In this study, we downloaded and analyzed UC-related datasets. Through differential analysis and Lasso regression, we identified six key cellular senescence genes, all of which were upregulated (TWIST1, IGFBP5, MME, IFNG, ME1, FOS). Using two algorithms, CIBERSORT and ssGSEA, we comprehensively investigated the immune infiltration landscape of these differentially expressed cellular senescence genes. Additionally, we constructed a diagnostic nomogram for UC using these six genes and validated its diagnostic capability and clinical application value. Furthermore, by assessing their expression levels, we evaluated their performance in predicting the response to biological therapies, particularly infliximab (IFX) or vedolizumab (VDZ). Finally, we validated the expression of these six genes in various cell types in UC single-cell data, and the results were consistent with the immune infiltration results. Twist1, a basic helix–loop–helix transcription factor, is implicated as a key mediator of epithelial–mesenchymal transition and is overexpressed in various cancer tissues^[157]27. TWIST1 regulates cellular senescence and energy metabolism in mesenchymal stem cells and plays a crucial role in epithelial-mesenchymal transition^[158]28. In osteoarthritis, the elevated expression of TWIST1 in cartilage exacerbates the catabolic response of human chondrocytes by upregulating MMP3 expression^[159]29. Furthermore, nuclear translocation of Twist1 represents a novel mechanism involved in the progression of osteoarthritis^[160]30. In IBD, the elevated expression of TWIST1 in intestinal tissues is associated with Th1-mediated intestinal immune responses, playing a crucial role in regulating the production of inflammatory cytokines by intestinal epithelial cells^[161]31,[162]32. Insulin-like growth factor (IGF) signaling plays a pivotal role in the regulation of cell growth, differentiation, apoptosis, and aging, with IGFBP5 being a significant member of the IGF axis^[163]33. IGFBP5 is induced in the cellular senescence process by the tumor suppressor p53^[164]34. In IBD, the heightened expression of IGFBP5 in inflammatory colonic tissues is linked to neutrophil gelatinase-associated lipocalin-mediated intestinal inflammation^[165]35,[166]36. Membrane metalloendopeptidase (MME) is a transmembrane glycoprotein with a distinctive extracellular protease domain capable of degrading various substrates^[167]37. MME can regulate the inflammatory response of adipocytes and insulin signaling^[168]38. Moreover, MME has been identified as a downstream effector of PI3K and can induce cellular senescence, a process that may be related to its glycosylation^[169]39. IFNG (IFN-γ) is a well-known pathway crucial in the immune response to inflammatory diseases^[170]40. IFNG activates innate immune responses, fostering the production of inflammatory cytokines and augmenting antigen presentation and natural killer cell function^[171]41. In IBD, IFNG contributes to disease pathogenesis by disrupting the vascular barrier through the upregulation of VE-cadherin^[172]42. The pro-inflammatory cytokine IFNG is essential for inducing cellular senescence^[173]43, and in turn, senescent cells can activate IFN signaling^[174]44. Malic enzyme 1 (ME1) is a cytoplasmic protein that catalyzes the conversion of malate to pyruvate, concurrently producing NADPH from NADP^[175]45. ME1 can also promote lipid synthesis and the onset of colorectal cancer^[176]46. In our analysis, although ME1 exhibited high expression in UC, it did not demonstrate a robust correlation in immune infiltration results and did not yield favorable outcomes in KM analysis after VDZ treatment (p > 0.01). FOS belongs to the cell cycle-related genes implicated in cell proliferation, growth, and differentiation, serving as a regulatory factor in cellular proliferation, differentiation, and transformation^[177]47. In specific contexts, the expression of the FOS gene is also associated with cellular senescence^[178]48,[179]49. In IBD, FOS mRNA levels are markedly expressed in UC but not in Crohn’s disease^[180]50. Likewise, high expression of FOS is observed in colonic tissues of DSS-induced colitis mice^[181]51. Diagnosing ulcerative colitis (UC) in patients can be challenging and often requires a combination of clinical history, endoscopic examination, pathological assessment, and imaging studies, which may lead to potential misdiagnosis^[182]1,[183]4,[184]23. The nomogram developed in this study demonstrated strong predictive capability and clinical utility for diagnosing UC, offering valuable guidance for IBD physicians in clinical decision-making. Biological therapies, such as vedolizumab and infliximab, are increasingly utilized in the treatment of moderate to severe UC due to their favorable therapeutic effects^[185]52,[186]53. Presently, biological agents are relatively expensive, with no definitive indicators available to accurately predict patient responsiveness to these drugs before use. Hence, we endeavored to utilize these six cellular senescence-related genes to forecast the response of UC patients to biological therapy. Intriguingly, our research results indicate that these six key genes are highly expressed in UC before treatment, and their post-treatment downregulation effectively predicts disease remission, demonstrating a robust correlation. Conversely, the expression changes of these six genes in UC patients who did not respond to treatment after therapy were not statistically significant. Additionally, single-cell RNA analysis further validated the expression of these six genes in UC. This study identified and analyzed features related to cellular senescence across multiple datasets for UC through bioinformatics analysis and machine learning. The alterations in expression observed in the intestinal mucosa of UC patients were confirmed in both a single-cell RNA dataset and a dataset derived from biological treatment for UC. As the era of an aging population approaches, aging is not only manifested as degenerative changes in the macroscopic world but also as dysregulation of cellular senescence in the microscopic world, which is closely associated with various diseases, especially those in the elderly. UC is a lifelong disease that cannot be completely cured. Under the stimulation of recurrent, chronic intestinal inflammation, the continuous disruption of the immune system in the intestine leads to abnormal expression of multiple genes controlling cellular senescence. By detecting the expression of these genes, we can predict the extent of cellular senescence changes in the UC intestine, which, to some extent, reflects local inflammation changes in the UC intestine and can predict the response to biologic therapy. In summary, our findings suggest that, unlike general population aging, intestinal aging in UC patients may occur prematurely. Through bioinformatics analysis, we have deepened our understanding of the role of cellular senescence in the development of UC and provided new avenues for exploring novel therapeutic targets for UC. Supplementary Information [187]Supplementary Information 1.^ (71.9KB, tsv) [188]Supplementary Information 2.^ (638.5KB, jpg) [189]Supplementary Information 3.^ (43.5KB, xls) [190]Supplementary Information 4.^ (101.2KB, r) [191]Supplementary Information 5.^ (3.1MB, tif) [192]Supplementary Information 6.^ (18.7KB, docx) [193]Supplementary Information 7.^ (53.3KB, xlsx) Acknowledgements