Abstract Background Pyroptosis, an emerging type of programmed cell death. The mechanisms of pyroptosis mainly include inflammasome-activated pyroptosis and non-inflammasome-activated pyroptosis. Multiple prognostic scoring systems that utilize pyroptosis-related gene expression have been validated as effective predictors of patient outcomes. But the relationship between pyroptosis and colorectal cancer remains unclear. This study has established a gene signature associated with pyroptosis to forecast the prognosis of CRC patients. Methods An analysis of 52 pyroptosis genes was conducted in both CRC and normal colorectal tissues, leading to the discovery of differentially expressed genes (DEGs). Core pyroptosis-related genes were identified using least absolute shrinkage and selection operator (LASSO) Cox regression to establish a prognostic risk score (PRS) for predicting CRC patient outcomes. The TCGA cohort was split into high-risk and low-risk groups based on the PRS, followed by Gene Ontology (GO) and KEGG pathway analyses. Additionally, differences in the enrichment scores of 16 immune cell types and the activity of 13 immune-related pathways were compared. The role of SPTBN5, a core pyroptosis-related gene, was validated through functional experiments on human colorectal adenocarcinoma cells (SW480). Results 40 differentially expressed genes were identified from 52 pyroptosis genes. A risk model was subsequently developed using 25 core pyroptosis-related genes identified through LASSO Cox regression analysis, and this model was validated in GEO cohorts. GO and KEGG pathway analyses showed that the DEGs are predominantly associated with mineral absorption, thyroid hormone synthesis, and pancreatic secretion. Functional experiments demonstrated that down-regulation of SPTBN5 expression through transfection led to significant decreases in the proliferation, migration, and clonogenicity of SW480 cells. Conclusion The PRS can identify high-risk CRC patient groups and predict patient prognosis. SPTBN5 may present a potential therapeutic target for CRC. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-024-01691-w. Keywords: Pyroptosis, CRC, Prognosis, SPTBN5 Introduction Colorectal cancer (CRC) is the second leading cause of cancer-related mortality in America [[30]1]. Treatment for individual CRC patients predominantly relies on prognostic factors validated by prior research. However, the efficacy of current prognostic markers remains limited. For instance, Tumor-Associated Trypsin Inhibitor(TATI), a trypsin inhibitor that functions mainly in the pancreas, has been recognized as an inflammatory marker and a standalone prognostic factor for CRC, with elevated preoperative s-TATI levels indicating a poorer prognosis [[31]2, [32]3]. A recent study revealed that TATI could not serve as an independent prognostic biomarker following multivariate analysis of patient cohorts [[33]4]. Furthermore, increased TATI levels may arise from tissue damage, inflammation, or the tumor itself [[34]5]. Pyroptosis, an emerging type of programmed cell death, typically instigated by inflammasomes and executed via gasdermin proteins. This process is characterized by cell swelling, membrane perforation, and the subsequent release of cellular contents. Under normal physiological conditions, pyroptosis serves a crucial role in host defense against pathogen infection. However, an excessive activation of pyroptosis can lead to uncontrolled and persistent inflammatory responses, thereby contributing to the onset of inflammatory diseases [[35]6]. Pyroptosis significantly influences the tumor microenvironment and is closely linked to both the initiation and progression of cancer. Research has shown that genes associated with pyroptosis can serve as standalone prognostic markers in various cancers, including ovarian cancer (OC) and gastric cancer (GC) [[36]7, [37]8]. In light of these findings, we conducted an extensive study to explore the role of pyroptosis-related genes and assess their prognostic value for patients with colorectal cancer (CRC). This was achieved by analyzing core pyroptosis-related genes and constructing a multi-gene risk model. Further investigation of Spectrin beta, non-erythrocytic 5 (SPTBN5), which was identified from a pool of 25 pyroptosis-related core genes using LASSO regression analysis, revealed its potential as a therapeutic target for CRC. Materials and methods Data RNA sequencing (RNA-seq) data along with clinical characteristics such as Sex, Age, TNM stage and survival information for 446 CRC patients were sourced from the TCGA database ([38]https://portal.gdc.cancer.gov/repository). Additionally, clinical data (Sex, Age, TNM stage and survival information) and RNA-seq data for the external validation cohort were retrieved from the GEO database ([39]https://www.ncbi.nlm.nih.gov/geo/, ID: [40]GSE40967). Identification of differentially expressed cell pyroptosis-related genes 52 pyroptosis genes, which were identified from the review, were listed in table S1 [[41]8]. GTEx data of 384 normal colon samples were analyzed to identify DEG between normal colon tissues and tumor tissues. To facilitate comparison, expression data in both datasets were standardized to fragments per kilobase million (FPKM). The 'limma' package was then used to identify DEGs (P-value < 0.05) (* for P < 0.05, ** for P < 0.01, *** for P < 0.001). The PPI network of the DEGs was generated by Search Tool for the Retrieval of Interacting Genes (STRING) version 11.0 ([42]https://string-db.org/). Construction and validation of a prognostic model based on pyroptosis-associated genes The correlation between each gene and the survival status of patients in the TCGA cohort was evaluated using Cox regression analysis to assess the potential value of pyroptosis-related genes in CRC prognosis. To enhance the reliability of our findings, we established a significance threshold of 0.2, which allowed us to identify 158 genes (Table S2) associated with survival for further analysis. Subsequently, we screened candidate genes using a least absolute shrinkage and selection operator (LASSO) Cox regression model (using the R package “glmnet”) and established a prognostic model. In the end, we identified 25 genes along with their corresponding coefficients (Supplementary Table S4). The penalty parameter (λ) was selected based on the minimum criterion. After applying centralization and standardization to the TCGA expression data (utilizing the 'scale' function in R), the risk score was computed using the formula: Risk Score = \ (\mathop {\ sum} olimits_i^25 {Xi \times Yi} \) (where X represents the gene coefficient and Y corresponds to the gene expression level). CRC patients in the TCGA cohort were stratified into low-risk and high-risk groups according to the median risk score, and survival times between the two groups were evaluated using Kaplan–Meier analysis. Principal component analysis (PCA) utilizing twenty-five gene features was conducted using the "prcomp" function from the R package "stats". Additionally, the R packages "survival", "survminer", and "timeROC" were employed to carry out a three-year ROC curve analysis. To further validate our results, we utilized a CRC cohort obtained from the GEO database ([43]GSE40967). In the [44]GSE40967 cohort, we standardized the expression levels of each pyroptosis-associated gene employing the "scale" function, and subsequently derived risk scores for these genes by applying the identical formula that was utilized for the TCGA cohort. Utilizing the median risk score derived from the TCGA cohort as a threshold, we stratified patients within the [45]GSE40967 cohort into low-risk and high-risk subgroups. Subsequently, we conducted a comparative analysis between these groups to verify the gene model. Independent prognostic analysis of risk scores Clinical data, encompassing age, tumor grade, and AJCC 8th edition staging information, alongside survival status, were extracted for individuals within the TCGA cohort. Subsequently, these variables were integrated with risk scores within the framework of both univariate and multivariate Cox regression models. This analytical approach aimed to comprehensively evaluate the predictive capabilities of risk scores, while also considering the potential impact of other clinical factors on patient outcomes. Functional enrichment analysis of DEGs between low-risk and high-risk groups In the TCGA cohort of CRC patients, we stratified individuals into two distinct subgroups based on the median risk score as a threshold. Subsequently, we applied rigorous criteria (|log2FC|≥ 1 and FDR < 0.05) to filter out differentially expressed genes (DEGs) that exhibited significant variations between the low-risk and high-risk groups. Utilizing the "clusterProfiler" package, we conducted comprehensive GO and KEGG analyses to investigate the functions and pathways of these DEGs. The immune cell infiltration scores were evaluated by ssGSEA calculation through "gsva" package, thereby evaluating the functionality and dynamics of immunological pathways. To obtain more information of the immune characteristics and biological differences of risk grouping in CRC. Cell culture and transfection SW480 cells were maintained in RPMI 1640 medium, under conditions of 37 °C and 5% CO2. Prior to transfection, these cells were plated onto a twelve-well dish and allowed to proliferate for 24 h until achieving a 70% confluency. Subsequently, transfection was performed by diluting lipo3000 and siRNA (SPTBN5) with 100 μl OPTI-MEM and left to stand at room temperature for 5 min. The siRNA and Lipo3000 were mixed and left to stand at room temperature for 20 min. Subsequently, the SW480 cells seeded in the twelve-well plate were washed with PBS and replenished with serum-deprived medium. Finally, the above mixture was added to a 12-well plate. Cellular immunofluorescence After culturing SW480 cells in a laser confocal dish for approximately 48 h, they were rinsed three times with PBS. Subsequently, fixation was performed at ambient temperature for a duration of 15 min, utilizing 1 ml of 4% paraformaldehyde as the fixative agent. Following this fixation step, the cells were again subjected to three rounds of PBS washing to eliminate the fixative. Lastly, permeabilization was carried out at ambient temperature for 15 min, employing a PBS solution that contained 0.2% Triton X. After another three washes with PBS, the cells were blocked at ambient temperature for 30 min with a PBS solution containing 4% BSA. Discard the blocking solution and add 100μL of SPTBN5 primary antibody diluted with blocking solution (ABMART, PA5987,1:1000), and incubate at 4℃ in a humid box for a night. After discarding the primary antibody, the cells were again subjected to three rounds of PBS washing. Under light-protected conditions, add the fluorescent secondary antibody solution ([46]A48254, diluted 1:250) of the same species as the primary antibody and incubate at ambient temperature for 1 h. After discarding the secondary antibody, the cells underwent a thorough washing procedure, involving three sequential rinses with PBS, to ensure the removal of any residual antibody. An appropriate amount of DAPI-containing mounting medium was added to cover the cells, and after 10 min, the stained results were scanned using an LSM900 laser confocal microscope. RT-qPCR The specified tissue or cellular material undergoes collection, subsequently undergoing total RNA extraction utilizing Trizol reagent (Beyotime brand) subsequent to pre-cooling at 4 °C. This RNA, derived from either cell or tissue samples, is then subjected to reverse transcription, with mRNA converted to cDNA. The reverse transcription system is prepared according to the kit's instructions, and reverse transcription is performed. After the reverse transcription is completed, the product is placed in a −80 ℃ refrigerator. mRNA is used for fluorescent quantitative PCR operation according to the user manual of SYBR company, PCR kit, which consists of a reaction system composed of 10 μL: 5 μL 2 × NovoStartSYBR QPCR SuperMix Plus, 1μL cDNA template, 0.2 μL upstream and downstream primers, 0.2 μL ROXI and 3.4 μL sterile water. The execution of the experimental protocol involves utilization of the ABI 7500 real-time PCR apparatus, sourced from ABI Corporation in the USA. As an internal control for mRNA quantification, β-actin serves as the reference gene, enabling the computation of the target gene's relative transcriptional level through application of the comparative quantification approach, specifically the 2-ΔΔCT methodology. The primer sequences are listed in Table S5. Cell scratch assay SW480 cells were planted onto six-well plates and subjected to transfection procedures. Following this, a clear demarcation line was created on the plate's surface utilizing a 200 μl pipette tip, and subsequently, the medium was replenished with 2% FBS. To assess the migratory capacity of these cells, microscopic images were captured at three distinct time points: 0 h, 12 h, and 48 h post-scratching. The analysis of cell migration was then conducted by quantifying the extent of scratch closure or healing area over this period. The area was quantified using the software ImageJ (Version 1.53 m). CCK8 SW480 cells, stratified into distinct groups, underwent trypsinization and were subsequently dispensed into a 96-well plate, ensuring an even distribution at a cellular density of 6 × 10^3 cells per well. Following a 48-h incubation period, 10 µl of CCK8 solution was introduced into each well, and the plates were further incubated at 37 °C. The OD values were measured at 6 h, 12 h, 24 h, 48 h, and 60 h respectively. Plate cloning experiment The processed SW480 cells were disseminated onto a 6-well plate and subjected to cultivation until discernible clusters of colonies became evident. Methanol was added, followed by fixation for 20 min. Then, 1% crystal violet staining solution was applied for 15 min. Afterwards, photographs were taken, and the number of cell clones formed was observed. Statistical analysis In order to assess the differential gene expression patterns between normal colon tissues and CRC tissues, a one-way ANOVA analysis was conducted. For the categorical variable comparisons, Pearson's chi-squared test was applied. Subsequently, the Kaplan–Meier survival curves, accompanied by two-sided log-rank tests, were employed to analyze the overall survival (OS) disparities among patient subgroups. To evaluate the independent prognostic significance of the risk models, both univariate and multivariate Cox regression analyses were undertaken. Additionally, when comparing immune cell infiltration levels and immune pathway activation status between two distinct groups, the Mann–Whitney U test was utilized. All statistical computations and analyses were executed utilizing the R statistical software package, version 4.0.2, adhering to rigorous academic standards. The overall flowchart is depicted as Fig. [47]1 and Supplementary Fig. 1. Fig. 1. [48]Fig. 1 [49]Open in a new tab Flowchart of all data analysis in the experiment Results Identification of DEGs between normal and tumor tissues After analyzing the GTEx and TCGA data of 384 normal and 446 tumor tissues, we conducted a comparative analysis of the expression levels of 52 pyroptosis genes, resulting in the identification of 40 differentially expressed genes (all P < 0.01). Among them, CHMP2A, CHMP6, NLRP2, NLRP7, GZMA, TP63, NLRP1, NLRC4, NLRP3, TIRAP, ELANE, PRKACA, IL18, CASP5, GSDMB, BAK1, CASP9, CHMP3, CHMP2B, CYCS, CASP3, IRF2, CHMP7, a total of 23 genes, were downregulated in tumor tissues, and BAX, GPX4, PJVK, NOD2, NOD1, PLCG1, CHMP4C, HMGB1, CASP8, GZMB, IL1A, IL1B, GSDMC, IL6, CASP4, GSDMA, TP53, a total of 17 genes, were upregulated in tumor tissues (Fig. [50]2A) (Blue represents low expression level, and red represents high expression level.). In order to delve deeper into the interplay among these genes associated with pyroptosis, we conducted a protein–protein interaction (PPI) analysis. The lowest interaction score was set to 0.9 (highest confidence), as shown in Fig. (Fig. [51]2B). The hub genes were NLRP3, NLRC4, CASP3, CYCS, CASP8, TP53, IL18, IL1B, CHMP3, CHMP2A, CHMP7 and CHMP5. The network depicting the relationships among all genes associated with pyroptosis is illustrated in Fig (Fig. [52]2C). Fig. 2. [53]Fig. 2 [54]Open in a new tab Expressions of the 40 pyroptosis-related genes and the interactions among them. A A heatmap illustrating the expression levels of these genes (green: low expression level; red: high expression level) of the pyroptosis-related genes between the normal (N, brilliant blue) and the tumour tissues (T, red). P values were indicated as: **P < 0.01; ***P < 0.001; B The PPI network demonstrating the interactions among pyroptosis-related genes (interaction score = 0.9); C The correlation network of pyroptosis-related genes is illustrated as follows: red lines indicate positive correlations, while blue lines denote negative correlations. The intensity of the colors reflects the strength of the associations Considering the tumor classification of DEGs To investigate the association between the manifestation of 40 DEGs pertaining to pyroptosis and the diverse subtypes of CRC, we conducted a consensus clustering approach encompassing the entire pool of 446 CRC patients sourced from the TCGA database. By varying the number of clusters (k) from 2 to 10, we observed that k = 2 yielded the highest intragroup correlation and the lowest intergroup correlation. The result suggests that 446 CRC patients could be well divided into two clusters based on 40 DEGs (Fig. [55]3A). The gene expression profiles and clinical features, including N, M, T stage of colorectal cancer, stage, age (≤ 60 or > 60 years old) and survival status (alive or dead) were visualized using a heatmap. Notably, significant differences were observed between the two clusters with respect to the N, M, and T stages of CRC and tumor stage (Fig. [56]3B). In addition, we conducted a comparative analysis of the OS duration between the two distinct clusters (Fig. [57]3C), and found obvious discrepancy (P = 0.042). Fig. 3. [58]Fig. 3 [59]Open in a new tab Tumor classification utilizing the expression profiles of pyroptosis-related DEGs. A Based on the consensus clustering matrix with k = 2, the 446 CRC patients were categorized into two distinct clusters; B Heatmap and the clinicopathologic characters of the two clusters classified by these DEGs; C Kaplan–Meier OS curves for the two clusters Establishment of a prognostic gene model within the TCGA cohort In total, 446 CRC samples were matched with the homologous patients who had comprehensive survival information. Genes associated with pyroptosis were selected by univariate Cox regression analysis. The results showed that 158 genes (Table S2) met the criteria of P < 0.2 and were reserved for ulteriorly investigating. Notably, 156 of these genes (Table S3) exhibited a positive correlation with elevated hazard ratios (HRs > 1), indicating an increased risk factor. In contrast, the remaining two genes (CCL22, FENDRR), stood out as protective factors, characterized by HRs less than 1 (Fig. [60]4A). A total of 25 core genes related to pyroptosis (IL20RB, MID2, IFITM10, LAMP5, CALB2, BANK1, ANGPTL4, CCL22, EGFL7, UPK3B, SPTBN5, TMPRSS11E, LINGO1, FENDRR, TRAF1, RNF207, ROBO3, TMEM88, GRP, SYNGR3, HOXC11, CHGB, HEYL, P2RX5, TOX2) were screened out by univariate Cox and LASSO Cox regression analysis. According to the optimal λ value, a total of 25 gene markers were constructed (Fig. [61]4B, C), and 446 CRC patients were divided into low and high risk subgroups by the median score calculated by the risk scoring formula (Fig. [62]4D). As verified by PCA, CRC patients with diverse risks were well separated into two groups (Fig. [63]4E). The high risk subgroup had more deaths and shorter survival times (Fig. [64]4F, High risk subgroups on the right of the dashed line). A notable disparity in OS was observed between the low risk subgroup and high risk subgroup (Fig. [65]4G, P < 0.001). The sensitivity and specificity of the prognostic model were assessed using time-dependent ROC analysis. The result shows that the AUC of the ROC curve was 0.800 for one year, 0.789 for two years, 0.777 for three years (Fig. [66]4H). Fig. 4. [67]Fig. 4 [68]Open in a new tab Development of a risk signature within the TCGA cohort. A Univariate Cox regression analysis was conducted to assess OS in relation to each pyroptosis-associated gene, and 158 genes with P < 0.2; B Through LASSO regression analysis, 25 OS-related genes were screened out; C Cross-validation was utilized to fine-tune parameter selection in the LASSO regression analysis; D Patients were categorized according to their risk scores; E PAC plot for CRC samples was generated based on the risk scores; F Survival status of all patients (low-risk population: on the left side of the dotted line; high-risk population: on the right side of the dotted line); G Kaplan–Meier survival curves were plotted to compare (OS) between patients in the high-risk and low-risk groups; H ROC curves were utilized to evaluate the predictive accuracy of the risk score External inspection of risk characteristics The validation set used 579 CRC patients from the GEO ([69]GSE40967). Prior to next analysis, the gene expression data was standardized using the "Scale" function. Utilizing the median risk score derived from the TCGA cohort as a benchmark, the GEO cohort was subsequently stratified, with 267 patients were categorized into the low risk group and the remaining 312 patients were allocated to the high risk group (Fig. [70]5A). PCA validation demonstrated that the CRC patients were well segregated into two distinct groups. (Fig. [71]5B). And it was found that low risk subgroup had fewer deaths and longer survival time (Fig. [72]5C, low risk subgroup on the left of the dashed line). Kaplan–Meier analysis showed that there was an obvious discrepancy in survival rate in the two subgroups (P = 0.002, Fig. [73]5D). The AUC of the ROC curve was 0.640 for one year, 0.607 for two years, 0.594 for three years. This result also indicates that our model exhibits robust predictive performance. (Fig. [74]5E). Fig. 5. [75]Fig. 5 [76]Open in a new tab Validation of risk models by GEO cohort. A Patients in the GEO cohort were categorized based on the median risk score derived from the TCGA cohort; B PCA plot for CRC samples; C The survival status of each patient was depicted with the low-risk population on the left side of the dotted line and the high-risk population on the right side; D Kaplan–Meier curves were generated to compare OS between the low-risk and high-risk groups; E Time-dependent ROC curves for CRC patients Assessment of the independent prognostic value of risk models Univariate and multivariate Cox regression analyses were employed to determine if the risk score obtained from the gene signature model could act as an independent prognostic factor. Univariate Cox regression analysis showed that in TCGA and GEO cohorts, the risk score was an independent factor predicting poor survival (HR = 4.349, 95% CI 3.288–5.752 and HR: 1.886, 95% CI 1.465–2.429) (Fig. [77]6A, C). Multivariate analysis further indicated that after adjusting for other confounding factors, the risk score remained an independent prognostic factor (HR: 3.452, 95% CI 2.532–4.708and HR: 1.374, 95% CI 1.091–1.731) (Fig. [78]6B, D) for CRC patients in both cohorts. Additionally, a heatmap of clinical characteristics was created for the TCGA cohort. (Fig. [79]6E). It was found that there were no significant differences in the distribution of N, M, T stages of colorectal cancer, Stage, age (≤ 60 or > 60 years), and survival status (alive or dead) between low-risk subgroup and high-risk subgroup. Fig. 6. [80]Fig. 6 [81]Open in a new tab Univariate and multivariate Cox regression analyses for the risk score. A Univariate analysis for the TCGA cohort; B Multivariate analysis for the TCGA cohort; C Univariate analysis for the GEO cohort; D Multivariate analysis for the GEO cohort; E A heatmap was generated to illustrate the relationships between clinicopathologic features and the risk groups (blue: low expression; red: high expression) (*P < 0.05) Gene function and pathway analysis based on risk models Using the "limma" R package (FDR < 0.05 and |log2FC|≥ 1) to screen DEGs in low and high risk subgroups of the TCGA cohort, to further explore differences in gene function and pathways between subgroups classified by the risk model.Then, GO enrichment analysis and KEGG pathway mapping were conducted using DEGs. It is shown that DEGs were primarily related to Mineral absorption, Thyroid hormone synthesis, Pancreatic secretion (Fig. [82]7A, B). Fig. 7. [83]Fig. 7 [84]Open in a new tab Gene function and pathway analysis based on the DEGs between the high-risk and low-risk groups in the TCGA cohort. A A bubble graph was used to illustrate GO enrichment analysis. In the graph, larger bubbles indicate a greater number of enriched genes, and deeper shades of red signify more pronounced differences. The q-value represents the adjusted p-value; B A bar plot was created to display the KEGG pathway enrichment analysis. In this graph, longer bars indicate a higher number of enriched genes, while deeper shades of red represent more pronounced differences Comparison of immune activity between high- and low-risk subgroups Based on functional analysis, the ssGSEA was used to further compare the enrichment scores of 16 distinct immune cell subsets and the activity levels of 13 pertinent immune-mediated pathways in the low risk subgroup and high risk subgroup in the TCGA and GEO cohorts. Compared with the low-risk subgroup, the levels of infiltration of immune cells were generally lower in the high-risk subgroup of TCGA cohort, especially for dendritic cells (DCs), induced dendritic cells (iDCs), helper T cells (Th1 and Th2), and regulatory T cells (Treg). Furthermore, the intensity of the T cell co-stimulation cascade and the type II IFN response mechanism, within the realm of immune signaling pathways, was markedly diminished in the high risk subgroup, as compared to their counterparts in the low risk subgroup (Fig. [85]8A, B). The same conclusion was drawn from the data analysis of the GEO cohort (Fig. [86]8C, D). Fig. 8. [87]Fig. 8 [88]Open in a new tab Immune cell and immune pathway analysis. A, B Comparison of enrichment scores for 16 types of immune cells and 13 immune-related pathways between the low-risk (blue box) and high-risk (red box) groups in the TCGA cohort; C, D Comparison of the tumour immunity between low- (blue box) and high-risk (red box) group in the GEO cohort. (P: ns not significant; *P < 0.05; **P < 0.01; ***P < 0.001) Functional verification of the SPTBN5 gene Among the 25 survival-related core genes screened out by univariate and multivariate Cox regression analyses, only the SPTBN5 gene has not been studied in tumors. Therefore, we performed a comprehensive study of the SPTBN5 gene. Firstly, we conducted a study on five colorectal cancer cell lines (SW480, LOVO, HT29, RKO and NCM460), and detected the expression level of SPTBN5 gene in them by RT-qPCR and cellular immunofluorescence. It was found that SPTBN5 was most significantly expressed in colon adenocarcinoma cells (SW480) (Fig. [89]9A, B). Three different siRNAs (siRNA1, siRNA2, siRNA3, primer sequences are listed in Table S5) were used to transfect SW480 cells, and the siRNA2 with the most significant transfection effect was selected for subsequent transfection experiments (Fig. [90]9C). Immunofluorescence results demonstrated (Fig. [91]10A) that the Ki67 protein content in the cells of the NC group was markedly elevated compared to the transfection group. Ki67, a pivotal marker of cellular proliferation, exhibits a profound functional linkage to the mitotic process, rendering it an indispensable component in the proliferation of cells. The higher the Ki67 content, the more active cell proliferation will be. In addition, the CCK8 experiment, plate clone experiment, and cell scratch experiment results of SW480 transfection group and NC group proved that knocking down SPTBN5 by transfection would reduce the cell's proliferation and migration ability (Fig. [92]10B–D). Fig. 9. [93]Fig. 9 [94]Open in a new tab The expression difference of SPTBN5 in different colorectal cancer cell lines and the transfection effect of different primers. A, B SPTBN5 mRNA expression levels in CRC cell lines; C Transfection effect of three SPTBN5 siRNA. (***P < 0.001) Fig. 10. [95]Fig. 10 [96]Open in a new tab Functional validation of SPTBN5. A Immunofluorescence assay; B CCK8 analysis of the proliferation of SW480 cells upon the transfections; C The effect of SPTBN5 down-regulation on migration function was evaluated by scratch tests, *P < 0.05; D Colony formation in SW480 cells was analyzed when SPTBN5 was knocked down compared with negative control, *P < 0.05 Discussion In our investigation, we delved into the mRNA expression profiles of 52 pyroptosis genes, comparing their levels in CRC tissues against those in normal counterparts, and revealing differential expression in 40 genes. A consensus clustering analysis on account of these DEGs successfully segregated CRC patients into low risk subgroup and high risk subgroup, which exhibited significant disparities in clinical characteristics and OS. In order to comprehensively assess the prognostic implications of the regulatory elements pertaining to pyroptosis, we formulated a risk prediction model, leveraging both univariate Cox and LASSO Cox regression analysis. The functional analysis suggested that the PRS derived from pyroptosis-related genes could act as an independent risk factor to identify high risk patient subgroup. DEGs between the two subgroups were associated with pathways such as mineral absorption, thyroid hormone synthesis, and pancreatic secretion. Furthermore, we performed an in-depth study of the SPTBN5 gene, one of the 25 core pyroptosis-related genes identified through LASSO regression analysis. Our findings revealed that downregulation of SPTBN5 by transfection significantly decreased the proliferation, migration and colony formation ability of SW480 cells. These consequences imply the potential of SPTBN5 as a viable therapeutic target in the CRC. Pyroptosis, a novel type of programmed cellular demise, holds a pivotal position within the intricate tumor microenvironment, intricately intertwined with the onset and advancement of neoplastic processes. On one hand, pyroptosis can instigate apoptosis in cancer cells, thereby inhibiting the initiation and progression of tumors. Conversely, pyroptosis, a type of pro-inflammatory death, can generate a microenvironment conducive to tumor cell proliferation, thereby promoting tumor growth [[97]9–[98]11]. Furthermore, the expression levels of genes associated with pyroptosis, their interplay, and their influence on cancer patient prognosis may exhibit variations across different cancers. For instance, research has indicated that the expression of the factors associated with pyroptosis is reduced in hepatocellular carcinoma (HCC) [[99]12]. In a study on HCC, the pyroptosis-related differentially expressed genes (DEGs) between HCC and non-tumor tissues were identified. Using Cox univariate and LASSO Cox regression analysis, a risk signature consisting of 10 genes was established. It was found that this risk signature had predictive value for the prognosis of HCC patients and was verified in the GEO database [[100]13]. Therefore, it is pertinent to develop a risk score related to cellular pyroptosis for CRC patients. In this study, we compiled a list of 52 publicly reported pyroptosis genes and conducted a systematic analysis of their expression patterns in both normal and tumor tissues. Forty significantly differentially expressed pyroptosis genes were identified. Notably, several of these genes, including NLRP1, NLRP3, CASP5, IL18, and IL1B, have been established as crucial components in the cell pyroptosis triggering pathway [[101]14]. For example, as the core molecule of the inflammasome, NLRP3 expression was silenced in CRC, thereby limiting gasdermin D (GSDMD)-mediated pyroptosis. NLRP1 and NLRP3 are involved in the assembly of the inflammasome complex, which cleave caspase-1 (CASP1) to mediate IL-18 production. IL-18 enhances epithelial barrier function and regeneration to resist CRC [[102]15, [103]16]. In our analysis results, the expression levels of NLRP1, NLRP3 and IL-18 genes in CRC patients were downregulated, which is consistent with the above research findings. Utilizing these 40 DEGS, we performed consensus clustering analysis to categorize CRC patients into low risk subgroup and high risk subgroup. Upon integrating clinical data analysis, we observed significant disparities in clinical features and OS between these two patient subgroups. This analytical approach bears resemblance to the research methodology employed in a study on ovarian cancer [[104]8]. Prognostic scoring system based on gene expression has been established as effective predictors of patient outcomes [[105]17, [106]18]. To further establish a pyroptosis-related gene signature that can accurately predict the prognosis of CRC patients, we screened 25 core genes related to cell pyroptosis through univariate Cox and LASSO Cox regression analysis. Then, based on the risk score formula, we calculated the PRS and separated all patients into low risk subgroup and high risk subgroup. Consistent with our hypothesis, patients in the low risk subgroup had a markedly superior survival rate. And the risk prediction model and its independent prognostic value were validated in GEO and TCGA cohorts. When using the GEO cohort for risk model validation, the AUC value of predicting survival was lower than that of the TCGA cohort (0640 vs 0800 at 1 year), which may be due to the different clinical or biological characteristics of the GEO cohort from the TCGA cohort and the smaller sample size in the GEO cohort. In addition, we analyzed DEGs between the two subgroups. Based on our GO and KEGG analysis findings, it was found that DEGs were predominantly associated with mineral absorption pathways, thyroid hormone synthesis pathways, and pancreatic secretion pathways. Mineral absorption pathways are intricately linked to hormone synthesis pathways, and they play a pivotal role in the numerous diseases associated with the human body. For instance, in the context of thyroid hormone synthesis, iodine, iron, and copper emerge as indispensable elements, whereas selenium and zinc facilitate the conversion of T4 to T3 [[107]19]. Iron, in particular, is an essential trace element that supports protein and enzyme activity across a broad spectrum of functions. Extensive epidemiological and experimental studys had shown that high levels of iron in the body have been correlated with the initiation, proliferation, and metastasis of malignancies [[108]20]. Iron is implicated in various tumor types, with colorectal cancer being the most prominent. Research indicates that excessive intestinal lumen iron can precipitate the onset and progression of colorectal cancer. Furthermore, prolonged exposure to iron compounds in drinking water can instigate and exacerbate ulcerative colitis and markedly elevate the prevalence of colorectal tumors. In mouse models, dietary heme has been shown to induce gut microbiota dysbiosis, lipid peroxidation, and exacerbated colitis, thereby fostering adenoma development [[109]21, [110]22]. Additionally, ferroptosis, a type of programmed cellular demise analogous to pyroptosis, has garnered increasing attention due to its potential association with various diseases [[111]23, [112]24]. Thyroid hormones are instrumental in modulating normal metabolism, development, and growth while also promoting cancer cell proliferation. Several studies have pointed out that hyperthyroidism and supplementation with thyroid hormones (TH) are predisposing risk factors for CRC [[113]25–[114]27]. The thyroid hormone receptor interacting protein 6 (TRIP6) belongs to the LIM family and serves as an adaptor protein, it is overexpressed in several tumor types, including colorectal cancer. Numerous studies have revealed that TRIP6 expression is markedly upregulated in CRC and correlates with tumor staging [[115]28, [116]29]. And the thyroid hormone T3 and its nuclear receptor TRα1 have certain effects on the phenotype of colon cancer stem cells and their response to chemotherapy. By analyzing the thyroid hormone status of patients, the chemotherapy treatment for CRC can be optimized [[117]30]. These findings align with our stratification of CRC patients based on PRS, wherein the high-risk subgroup exhibited markedly elevated tumor malignancy and reduced OS. This underscores the validity of the constructed risk model for colorectal cancer pyroptosis-related genes. Moreover, we hypothesize that the augmentation of the mineral absorption pathway underlies the enhancement of the thyroid hormone synthesis pathway, subsequently influencing the onset and progression of CRC. Consequently, these two signaling pathways may represent novel avenues for CRC treatment research. Furthermore, 25 of the core genes linked to pyroptosis have been proven to contribute to tumor progression. For example, IL20RB is involved in the JAK/STAT pathway that fosters tumor progression, and its elevated expression correlates with a decreased overall survival rate in patients [[118]31]. MID2, a ubiquitin-conjugating E2 enzyme, has been identified as a new interaction companion of breast cancer 1 (BRCA1). Elevated expression level of MID2 is intimately correlated with advanced clinical stages of breast cancer, as well as the T, N, and M staging, with all associations reaching statistical significance (P < 0.05) [[119]32]. CALB2 facilitates the activation of the TRPV2-Ca2 + -ERK1/2 signaling pathway, which induces metastasis in HCC cells. Additionally, research suggests that CALB2 could be a underlying target for gemcitabine (GEM) therapy in CRC [[120]33, [121]34]. SPTBN5 is a core gene associated with pyroptosis. Mammalian β-spectrin comprises two α subunits and five β subunits. The αI subunit is genetically determined by SPTA1, whereas the αII subunit is encoded by SPTAN1. And the βI-V spectrin subunits are encoded by SPTB, SPTBN1, SPTBN2, SPTBN4, and SPTBN5. The occurrence of pathological mutations within either the spectrin α or β subunits has the potential to precipitate a diverse spectrum of illnesses, encompassing conditions such as hemolytic anemia, neurodegenerative disorders, ataxia, cardiac ailments, and even malignancies [[122]35]. For instance, SPTBN1, the most highly expressed member of the β-spectrin within the brain, is crucial for the structural integrity of neurons. It plays significant roles in axon stability and transport, synaptic transmission, regulation of neurite outgrowth during synaptogenesis, and proper myelination of Schwann cells. Studies have shown that mice lacking SPTBN1 in all neural progenitor cells exhibit early postnatal death, impaired distal cortical and cerebellar connections, spontaneous seizures, and motor coordination defects [[123]36, [124]37]. Loss of SPTBN1 activates Wnt signaling and promotes tumorigenesis and invasion in HCC cells in our body [[125]38]. Furthermore, SPTBN1 is implicated in the onset and progression of various cancers as per recent reviews [[126]37, [127]39]. SPTBN2, predominantly expressed in cerebellar Purkinje cells, plays a crucial role in the maintenance of dendritic architecture. It also facilitates the trafficking and stabilization of various membrane proteins [[128]40]. Heterozygous mutations in SPTBN2 can cause autosomal dominant spinocerebellar ataxia 5 (SCA5), while homozygous mutations are thought to cause autosomal recessive spinocerebellar ataxia 14 (SCAR14). SPTBN2 is also implicated in various cancers. For instance, it exhibits a positive correlation with poor prognosis in lung adenocarcinoma and ovarian cancer. Furthermore, research has indicated that the N6-methyladenosine (M6A) modification of SPTBN2 is associated with tumor growth and survival in CRC patients [[129]41–[130]43]. SPTBN4 is predominantly localized to the axon initial segments (AISs) and nodes of Ranvier, and assuming a pivotal function in the sustenance of membrane potential stability. It also initiates, propagates, and regulates action potentials by interacting with ankyrin-G, which clusters KCNQ2/3-potassium channels and NaV-sodium channels [[131]44]. Homozygous or compound heterozygous variants in SPTBN4 can lead to neurodevelopmental disorders, characterized by hypotonia, neuropathy, and deafness [[132]35]. Within the β-spectrin family, SPTBN5 is the only gene that has not been extensively investigated in relation to diseases. Consequently, we conducted a comprehensive study on the SPTBN5 gene. Our investigation uncovered a conspicuously elevated expression of SPTBN5 in human SW480 cells, which surpassed both the levels observed in non-neoplastic colon tissue cells and those documented in other colorectal cancer cell lines. As a result, we selected SW480 cells for the functional validation of the SPTBN5 gene. The findings indicated that following transfection and knockdown of SPTBN5, there was a significant reduction in the proliferation, migration, and clonogenicity of SW480 cells. Therefore, we propose that SPTBN5 could be one of the potential therapeutic targets for CRC. In conclusion, our study suggests a strong correlation between CRC and pyroptosis. The multi-gene risk model we developed, centered on core genes associated with pyroptosis, demonstrates potential in predicting the prognosis of CRC patients and could inform personalized treatment strategies. Moreover, we identified the SPTBN5 gene as a potential therapeutic target for CRC, which could guide the development of novel therapies. However, further studies are needed to validate these findings in larger, independent cohorts, and to explore the underlying mechanisms of pyroptosis in CRC progression. Future research should also investigate the clinical applicability of the multi-gene model in optimizing treatment plans for CRC patients. Supplementary Information [133]12672_2024_1691_MOESM1_ESM.tif^ (2.2MB, tif) Supplementary material 1: Fig.1 Graphic abstract of this study. [134]Supplementary material 2.^ (8.9KB, xlsx) [135]Supplementary material 3.^ (19.2KB, xlsx) [136]Supplementary material 4.^ (19.1KB, xlsx) [137]Supplementary material 5.^ (9.4KB, xlsx) [138]Supplementary material 6.^ (8.7KB, xlsx) Author contributions GYL: writing-original draft, conceptualization; PYW: methodology, data curation, investigation, visualization; XNF: software; YXL: supervision and editing. Funding This research was funded by Anhui Provincial Health Commission Provincial Financial Support for Youth Programs (Guangyao Li, Grant No. AHWJ2023A30159). Data availability No datasets were generated or analysed during the current study. Declarations Ethics approval and consent to participate The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of The Second People’s Hospital of Wuhu (2023-KY-010). The written informed consent was obtained from each patient. Consent for publication All authors approved the content and submission. Competing interests The authors declare no competing interests. Footnotes Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Guangyao Li and Pingyu Wang have contributed equally to this work. References