Abstract Early diagnosis and disease management based on risk stratification have a very positive impact on colon adenocarcinoma (COAD) prognosis. It is of positive significance to further explore risk stratification of COAD patients and identify predictive molecular biomarkers. PANoptosis is defined as a form of inflammatory cell death regulated by PANoptosome, with common features of pyroptosis, apoptosis and necroptosis. The role of PANoptosis in COAD has not been fully studied. In this study, we analyzed significant differences in the expression of PANoptosis-related gene (PRG) features in COAD. Subsequently, the PANoptosis associated lncRNAs (PALs) associated with PRGs were analyzed by LASSO algorithm and multivariate Cox analysis, and PALs related to the prognosis of COAD were selected. Based on the expression patterns of prognostic PAL features, we performed unsupervised consensus cluster analysis to categorize COAD samples into distinct PAL molecular subtypes and investigate their associated immune infiltration characteristics. We subsequently constructed PAL score model based on prognostic characteristics and verified its independent prognostic value for COAD. The nomogram diagnostic model was established to confirm the prognostic value of PAL scoring system again. Pathway enrichment analysis, somatic mutation profiling, and drug sensitivity analysis were employed to comprehensively assess the clinical value of the PAL score. Additionally, qRT-PCR was used to further validate the abnormal expression of the selected targets in COAD. Our results provide a new idea for clinical risk stratification and new evidence for the role of PANoptosis in COAD. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-01838-3. Keywords: Colon adenocarcinoma, PANoptosis, lncRNA, Immune infiltration, Prognostic Introduction Despite advances in disease management, colon adenocarcinoma (COAD) remains one of the leading causes of death worldwide [[36]1]. Early diagnosis and disease management based on risk stratification had a very positive impact on COAD prognosis [[37]2, [38]3]. Therefore, developing accurate risk assessment models and identifying predictive molecular biomarkers is urgently needed for COAD patients [[39]4]. Long non-coding RNAs (lncRNAs) are involved in a variety of biological processes [[40]5]. LncRNAs can interact with RNA, DNA, and proteins to form complexes, thereby regulating gene expression through various mechanisms, such as modulating transcription, mRNA stability, and translation [[41]6, [42]7]. In addition, lncRNAs can also regulate the level of chemical reactions by acting as scaffolds or decoys for proteins or RNA, or adjust gene expression by influencing the structure of chromatin [[43]5, [44]8]. LncRNAs also play a crucial role in the occurrence and progression of COAD [[45]9]. In COAD, multiple biological functions including cell proliferation, apoptosis, cell metastasis and invasion, cell cycle, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), and drug resistance have been reported to be associated with lncRNAs [[46]10]. PANoptosis, a unique form of programmed cell death (PCD) discovered in recent years, is defined as an inflammatory cell death regulated by the PANoptosome. It shares key characteristics of pyroptosis, apoptosis, and necroptosis [[47]11]. The key components of PANoptosome include RIPK1, ASC, RIPK3, CASP6, ZBP1 and CASP1 [[48]12]. PANoptosome assembly is mediated by key molecular motifs collectively known as death fold domains, and defects in its components have been implicated in a range of human diseases, including carcinogenesis process [[49]13]. The potential prognostic value of PANoptosis for COAD has been reported [[50]14]. PANoptosis is regulated by IRF1 to prevent colorectal cancer [[51]15]. However, the role of PANoptosis in COAD is not fully studied. In this study, we screened PANoptosis associated lncRNAs (PALs) that were associated with the prognosis of COAD. Based on the expression patterns of prognostic PAL features, COAD samples were classified into distinct PAL molecular subtypes, which were then analyzed for variations in immune infiltration characteristics. Then we constructed a PAL scoring model based on the prognostic characteristics, and verified the prognostic value of the PAL scoring system by establishing a nomogram diagnostic model. Pathway enrichment analysis, somatic mutation characteristics and drug sensitivity analysis were used to evaluate the clinical value of PAL score. qRT-PCR was used to further validate the abnormal expression of the selected target in COAD. Our results provide a new idea for clinical risk stratification and a new basis for the role of PANoptosis in COAD. Materials and methods Transcriptome data collection and preprocessing In this study, transcriptome data for COAD were obtained from the publicly accessible The Cancer Genome Atlas (TCGA) database. Using the Perl programming language, we extracted and integrated transcriptome files for each sample. Gene annotation for each sample was performed using the Ensemble human genome browser GRCh38.p13 annotation file within the Perl framework. Concurrently, clinical baseline data for COAD samples were extracted from the TCGA database using Perl. After excluding samples lacking clinical survival information, a total of 41 normal samples and 446 COAD samples with complete clinical baseline data were collected for subsequent analyses. Copy number variation (CNV) frequency files were downloaded from the UCSC Xena database ([52]https://xenabrowser.net/datapages/). The STRING database was utilized to predict the interaction network among genes. Identification and prognostic value analysis of lncRNAs associated with PANoptosis Based on previously reported literature, we collected a panel of 14 PANoptosis-related genes (PRGs) (Supplementary Table 1) [[53]16]. Using the Pearson algorithm, we calculated the potential associations between these 14 PRGs and lncRNAs, identifying PANoptosis-associated lncRNAs (PALs). A filtering threshold was set at |r|> 0.5 and p < 0.05. Utilizing the "survival" R package, we performed single-factor Cox analysis to evaluate the association of PALs with clinical prognosis in COAD and computed hazard ratios (HR) and p-values for each PAL variable. Subsequently, employing the "glmnet" R package, we constructed a LASSO regression model to further identify key PAL variables. Furthermore, using multi-factor Cox analysis, we identified PALs with independent prognostic value and calculated the risk scores for each variable. The potential associations between PALs and the 14 PRGs were evaluated using the "ggplot2" R package. Identification of unsupervised molecular subtypes of PALs Based on PAL expression features identified with independent prognostic value, we employed the "ConsensusClusterPlus" script to explore molecular subtype characteristics of PALs in COAD samples. Using the k-means clustering algorithm, COAD samples were classified into subgroups ranging from k = 2 to 9, and the optimal model parameters were determined to define PAL molecular subgroups. The "ggplot2" script was used to visualize an unsupervised Principal Component Analysis (PCA) model, examining distribution patterns among different molecular subgroups. Clinical survival curves for PAL molecular subgroups were plotted using the "survival" script. Assessment of immune infiltration status features Based on the transcriptomic features of samples, we utilized the "ESTIMATE" script to calculate immune scores, stromal scores, ESTIMATE scores, and tumor purity for each sample. These four metrics were used to evaluate the immune infiltration status of the samples. Additionally, using the "GSVA" script and known markers for 23 immune cell types, we conducted ssGSEA analysis to quantitatively assess the infiltration proportions of these 23 immune cell types in each sample. Using the Pearson correlation algorithm, we employed the "ggplot2" script to compute correlations between independent prognostic variables of PALs and the 23 immune cell types. Construction of PAL scoring system model and independent prognostic analysis Based on the independent prognostic features and risk coefficients of PAL expression, we calculated PAL scores for each COAD sample using the formula: PAL Score = Prognostic Variable Expression Feature * Risk Coefficient. The "caret" script was used to divide COAD samples into training and validation sets in a 7:3 ratio based on PAL’s independent prognostic features. The samples were then stratified into two distinct PAL score subgroups using the optimal cutoff value derived from clinical survival prognosis. By combining clinical baseline data with PAL scores, we conducted univariate and multivariate Cox analyses using the "survival" script to evaluate the independent prognostic value of various clinical pathological indicators and PAL scores. ROC curves for 1-year, 3-year, and 5-year outcomes for different PAL score cohorts were plotted using the "survivalROC" script to assess their diagnostic performance. A nomogram diagnostic model was constructed based on clinical pathological variables and PAL score models using the "rms" script. The consistency between the nomogram diagnostic model and actual survival rates was visualized with the "regplot" script. The concordance index between PAL scores and clinical pathological variables was assessed using "pec," "rms," "dplyr," and "survival" scripts. Finally, decision curves for the nomogram diagnostic model and other variables were plotted using the "ggDCA" R script to evaluate the precision of each variable. Analysis of differential gene expression and somatic mutation features in PAL score subgroups Using a differential threshold of |fold change|> 2 and p < 0.05, we applied the "limma" script to identify genes differentially expressed between PAL score subgroups. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were then performed on these differentially expressed genes using the "clusterProfiler" script to explore potential molecular mechanisms. Somatic mutation data (MAF files) for COAD were retrieved from the TCGA database and processed with Perl. The "maftools" R script was used to analyze somatic mutation features across the PAL score subgroups. Immune therapy response data for COAD samples, including PD1 and CTLA4 responses, were sourced from the TICA database, and the "limma" script was utilized to evaluate the response of PAL subgroups to immune therapies. Additionally, using data from the GDSC database, we employed the "pRRophetic" R package to predict potential responses to small molecule targeted drugs in different PAL score subgroups. Cell culture NCM460 cells (human normal colon epithelial cells) were obtained from InCell Corporation (San Antonio, TX, USA). Cells were cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F-12, Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS, Gibco), 100 U/mL penicillin, and 100 μg/mL streptomycin (Gibco). Cells were maintained in a humidified incubator at 37 °C with 5% CO₂. The culture medium was changed every 2–3 days, and cells were subcultured at a ratio of 1:3 when reaching 80–90% confluence. HCT116 cells (human colon cancer cell line) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). Cells were cultured in RPMI 1640 medium (Gibco) supplemented with 10% fetal bovine serum (FBS, Gibco), 100 U/mL penicillin, and 100 μg/mL streptomycin (Gibco). Cells were maintained in a humidified incubator at 37 °C with 5% CO₂. The culture medium was changed every 2–3 days, and cells were cultured at a ratio of 1:4 when reaching 80–90% confluence. Quantitative real-time PCR (qRT-PCR) analysis qRT-PCR was performed to analyze gene expression levels. Total RNA was isolated from the cultured cells using a commercially available RNA extraction kit following the manufacturer's instructions. The concentration and purity of RNA were determined using a spectrophotometer. Subsequently, cDNA synthesis was carried out using a reverse transcription kit according to the manufacturer's protocol. The qRT-PCR reactions were prepared using a master mix that included cDNA, gene-specific primers, and SYBR Green PCR Master Mix. The reactions were conducted in triplicate on a real-time PCR machine, following the appropriate cycling conditions. GAPDH or β-actin was used as an internal control for normalization. Relative gene expression levels were calculated using the 2^-ΔΔCt method. Primers pairs are shown in Supplementary Table 2. All experiments were conducted in accordance with standard laboratory protocols and best practices. Data analysis In this study, all data preprocessing and analyses were conducted using R version 4.1.0 and Perl. Statistical differences between two groups were assessed using the Wilcoxon rank-sum test. For comparisons involving more than two groups, statistical differences were evaluated using ANOVA. Pearson correlation analysis was employed to assess correlation coefficients between variables. Cellular experimental data were statistically analyzed using GraphPad Prism 9.0 software (GraphPad Software, San Diego, CA, USA). Experimental data are presented as mean ± standard deviation (Mean ± SD). Each experiment was performed independently at least three times. A p-value less than 0.05 was considered statistically significant, with significance levels denoted as *P < 0.05, **P < 0.01, and ***P < 0.001. All p-values were two-tailed. Results Expression and mutation landscape features of PRGs in COAD To elucidate the potential roles of PRGs in COAD, we utilized the "limma" script to analyze the differential expression of 14 PRGs between normal and tumor tissues. Differential expression analysis revealed significant differences in 10 out of the 14 PRGs. Specifically, ZBP1, NLRP3, RIPK1, RIPK3, and TNFAIP3 were markedly downregulated in tumor tissues, while CASP8, FADD, MAP3K7, RBCK1, and PSTPIP2 were significantly overexpressed (Fig. [54]1A). Copy number variation (CNV) analysis of the PRG signature indicated significant amplifications in ZBP1, PYCARD, NLRP3, and TNFAIP3, whereas RBCK1 exhibited notable copy number deletions in COAD (Fig. [55]1B). Mutation landscape analysis revealed that a substantial proportion of PRGs harbored mutations in COAD, with mutation rates of 5% for NLRP3, 4% for CASP8, 3% for TNFAIP3, and 3% for RNF31 (Fig. [56]1C). Analysis of protein–protein interaction (PPI) networks depicted significant interactions among the 14 PRGs (Fig. [57]1D). These findings highlight notable expression differences and genomic alterations in the PRG signature in COAD, emphasizing their potential roles in the carcinogenesis process. Fig. 1. [58]Fig. 1 [59]Open in a new tab Differential Expression Analysis and Mutation Landscape Features of PRG Signature. A Differential expression analysis of PRGs between normal and tumor tissues in COAD. B Copy number variation analysis of PRG signature in COAD. C Mutation landscape features of PRG signature in COAD. D Protein–protein interaction (PPI) network diagram illustrating potential interactions among the 14 PRG signatures Identification and prognostic analysis of PANoptosis-associated lncRNA Using Pearson correlation analysis, we identified PALs signature significantly associated with the PRGs and evaluated their correlation with clinical prognosis in COAD. Setting a correlation threshold (r) greater than 0.5, we identified and obtained 993 PALs associated with PRGs. The Sankey diagram in Fig. [60]2A illustrates the mutual associations between the PRG signature and PALs. By integrating PAL expression features with clinical survival baseline data in COAD, we evaluated the prognostic value of PALs. Univariate Cox analysis identified 35 PALs significantly associated with clinical prognosis in COAD, including one protective factor and 34 risk factors (Fig. [61]2B). Using LASSO algorithm analysis, we further narrowed down the prognostic PAL variables and identified 8 feature variables (Fig. [62]2C). Subsequently, through multi-factor Cox analysis, we evaluated the independent predictive value of these 8 feature variables for predicting clinical prognosis in COAD and identified 6 PAL variables with independent prognostic value. A correlation heatmap (Fig. [63]2D) showed significant associations between these 6 independent prognostic PALs and the 14 PRGs. Fig. 2. [64]Fig. 2 [65]Open in a new tab Identification of PANoptosis-Related lncRNAs in COAD. A Sankey diagram illustrating the mutual associations between PALs and the PRG signature. Correlation threshold set at |r|> 0.5. B Univariate Cox analysis evaluating the prognostic value of PALs in COAD. C LASSO analysis used to select prognostic PAL variables. D Correlation analysis showing the relationship between independent prognostic PAL variables and the PRG signature PAL molecular subtype characterization based on prognostic PAL signatures Based on the prognostic PAL signature's expression features, we performed unsupervised consensus clustering analysis to classify COAD samples into distinct PAL molecular subtypes. According to the optimal clustering parameters, COAD samples were categorized into three PAL subgroups: PAL Subgroup A with 146 samples, PAL Subgroup B with 204 samples, and PAL Subgroup C with 96 samples (Fig. [66]3A). PCA plots demonstrated significant separation among the three PAL molecular subgroups, indicating distinct characteristics between them (Fig. [67]3B). Clinical prognosis analysis indicated that PAL Subgroup B had poorer clinical outcomes compared to Subgroups A and C, suggesting that Subgroups A and C may offer potentially better clinical survival benefits (Fig. [68]3C). Using ESTIMATE algorithm, we evaluated the immune infiltration status characteristics among different PAL molecular subgroups. The assessment showed that in PAL Subgroup C, associated with the best prognosis, stromal score and ESTIMATE score were significantly lower, while tumor purity was markedly higher, indicating a lower immune infiltration status potentially associated with clinical survival benefits in COAD (Fig. [69]3D–G). Using ssGSEA algorithm, we quantitatively analyzed the relative abundance of 23 immune cell infiltrates within PAL subgroups. Notably, in the PAL Subgroup C with the best clinical prognosis, a significant downregulation was observed in the proportions of most immune cell infiltrates, such as activated B cells, activated CD8 T cells, activated dendritic cells, underscoring the close association between immune activation status and adverse prognosis in COAD (Fig. [70]3H). Visualization heatmap results revealed differential expression patterns of the 6 independent prognostic PALs across different PAL molecular subgroups and clinical features (F[71]ig. [72]3I). Based on these findings, we conclude that the independent prognostic features derived from the PAL score accurately classify COAD samples into distinct molecular subtypes, with immune activation status potentially playing a key role in the adverse prognosis of COAD. Fig. 3. [73]Fig. 3 [74]Open in a new tab Molecular Subtype Features and Immune Infiltration Analysis of PAL in COAD. A Consensus clustering analysis of PAL molecular subtypes based on prognostic features. B PCA plot analysis of PAL molecular subgroups. C Clinical survival outcome analysis of PAL molecular subtypes. D–G ESTIMATE algorithm revealing the immune infiltration status of PAL molecular subgroups. H Quantitative analysis of 23 immune cell infiltrates in PAL molecular subtypes using ssGSEA. I Differential expression patterns of PAL prognostic features across different clinical features and PAL subgroups Construction of PAL score models based on prognostic features We developed a systematic PAL score model based on prognostic expression features of PALs, calculating PAL scores for each COAD sample. Using optimal clinical survival cutoffs, we stratified COAD samples into two PAL score subgroups across the complete, training, and validation sets of the PAL score model (Fig. [75]4A–C). Survival analysis across the three independent PAL score models revealed significantly better overall survival rates in the low PAL score subgroup compared to the high PAL score subgroup, indicating a potential association of high PAL scores with adverse prognosis in COAD (Fig. [76]4D–F). PCA analysis based on prognostic expression features of PAL successfully differentiated PAL score subgroups, emphasizing their independence and variability (F[77]ig. [78]4G–I). These findings suggest that the PAL score model, based on independent prognostic features of PAL, accurately predicts clinical outcomes in COAD, facilitating stratification of COAD samples into distinct risk subgroups. Fig. 4. [79]Fig. 4 [80]Open in a new tab Establishment and Independence Analysis of PAL Score System Models in COAD. A–C Construction and stratification of PAL score subgroups in the complete PAL score model, training set PAL score model, and validation set PAL score model. D–F Clinical prognostic outcome analysis of COAD across three independent PAL score models. G–I Unsupervised PCA analysis based on prognostic features of PAL in COAD Analysis of the independent prognostic value of the PAL score system By integrating the PAL score system with clinical-pathological features, we were able to further assess the independence and stability of PAL scores in predicting clinical outcomes in COAD. Through univariate and multivariate Cox regression analyses, incorporating both clinical-pathological variables and PAL scores, we calculated hazard ratios (HR) to investigate their potential associations with COAD prognosis. As depicted in Fig. [81]5A–C, across the three independent PAL score cohorts, the PAL score model emerged as a significant risk factor associated with adverse prognosis in COAD (HR > 1, P < 0.05), indicating that the PAL score system is an independent prognostic factor for COAD. Time-dependent ROC curve analysis demonstrated that in the complete PAL score model, the area under the curve (AUC) values for 1-year, 3-year, and 5-year survival were 0.692, 0.731, and 0.723, respectively. In the training set PAL score model, the corresponding AUCs were 0.701, 0.751, and 0.764, and in the validation set PAL score model, they were 0.671, 0.686, and 0.616 (Fig. [82]5D–F). Fig. 5. [83]Fig. 5 [84]Open in a new tab Assessment of the Independent Prognostic Value of PAL Score Models in Various Independent Cohorts. A–C Evaluation of the independence prediction of clinicopathological features and PAL score models across different independent cohorts. D–F Time-dependent ROC curve evaluation across three independent cohorts Development of a nomogram diagnostic model based on the PAL scoring model Given the independent prognostic value of the PAL scoring system and clinicopathological features in COAD prognosis, we developed a nomogram diagnostic model based on various clinical baseline variables and PAL scores to assess clinical survival probabilities at 1 year, 3 years, and 5 years (Fig. [85]6A). ROC curve analysis revealed that the nomogram diagnostic model achieved an AUC of 0.822, significantly outperforming standalone PAL scores and other pathological features, underscoring its superior predictive capability compared to conventional clinical indicators (Fig. [86]6B). Additionally, calibration curve analysis showed a good correlation between the predicted and observed survival probabilities across various time points (Fig. [87]6C). The concordance index curve analysis demonstrated that the PAL scoring system had superior prognostic accuracy compared to other pathological features (Fig. [88]6D). Decision curve analysis revealed that the nomogram diagnostic model offered more accurate survival predictions for COAD than both the PAL scores and other clinical-pathological features (Fig. [89]6E). These findings suggest that the nomogram diagnostic model, incorporating PAL scores and clinicopathological features, effectively evaluates survival probabilities at different time points in COAD, highlighting its substantial potential in prognosticating COAD survival outcomes. Fig. 6. [90]Fig. 6 [91]Open in a new tab Development of the nomogram diagnostic model based on the PAL scoring system and clinicopathological features. A Construction of the nomogram diagnostic model. B Assessment of diagnostic performance of the nomogram diagnostic model, PAL scoring system, and pathological features. C Calibration curve analysis. D Concordance index analysis between PAL scores and clinicopathological features. E Decision curve analysis Analysis of immune infiltration characteristics in PAL score subgroups We further assessed the immune infiltration profiles across PAL score subgroups to explore potential mechanisms underlying the clinical survival differences. ESTIMATE analysis revealed that, in the high PAL score subgroup, immune score, stromal score, and ESTIMATE score were significantly higher, while tumor purity was notably lower (Fig. [92]7A–D). These findings further suggest a close association between heightened immune activation and poor prognosis in COAD. Additionally, quantitative results from ssGSEA demonstrated significant upregulation of immune cell populations in the high PAL score subgroup, including MDSCs, macrophages, regulatory T cells, among others (Fig. [93]7E). Using Pearson correlation analysis, we evaluated the relationships between PAL prognostic signatures and immune infiltrating cells, revealing significant positive correlations of MYOSLID and ATP2B1-AS1 with 23 immune infiltrating cells, whereas [94]AL137782.1, [95]AC019118.1, FLJ21408, and [96]AC092944.1 showed significant negative correlations with immune infiltration status (Fig. [97]7F). In summary, based on these results, we elucidated the immune infiltration characteristics among PAL score subgroups, providing preliminary insights into the association between adverse clinical outcomes in COAD and immune activation status. Fig. 7. [98]Fig. 7 [99]Open in a new tab Association Analysis of PAL Score Subgroups with Immune Infiltration Status. A–D Assessment of immune infiltration status based on ESTIMATE algorithm. E Quantitative analysis of 23 immune infiltrating cells using ssGSEA algorithm. F Correlation analysis between PAL prognostic signatures and immune infiltration features Characterization of potential regulatory mechanisms and somatic mutations in PAL scoring subgroups We assessed gene expression differences between PAL score subgroups using a differential threshold of |FC|> 2 and p < 0.05, exploring potential regulatory mechanisms (Fig. [100]8A). GO analysis of differentially expressed genes revealed biological functions related to regulation of cellular response to growth factor stimulus, collagen-containing extracellular matrix, and extracellular matrix structural constituent (Fig. [101]8B). KEGG enrichment analysis identified several signaling pathways, including cytoskeleton regulation in muscle cells, malaria, dilated cardiomyopathy, and vascular smooth muscle contraction, as potential key regulatory mechanisms in COAD (Fig. [102]8C). Somatic mutation analysis revealed significantly higher mutation frequencies in many somatic genes in the high PAL score subgroup, such as APC, TP53, and TTN (Fig. [103]8D, E). IPS immune response evaluation suggested that the low PAL score subgroup showed better responses to CTLA4 or PD1 immune therapies (Fig. [104]8F). Furthermore, our predictions identified potential beneficial anti-tumor drugs for PAL score subgroups; IC50 results indicated significantly lower scores for Bortezomib, CP466722, Crizotinib, and MG-132 in the high PAL score subgroup, suggesting potential sensitivity to these treatments (Fig. [105]8G). In summary, our findings provide preliminary insights into the potential regulatory mechanisms among PAL score subgroups, predict responses to immune and targeted therapies, and offer new perspectives for future personalized precision medicine approaches. Fig. 8. [106]Fig. 8 [107]Open in a new tab Analysis of Potential Regulatory Mechanisms and Immune Therapy Response in PAL Score Subgroups. A Differential expression analysis of genes between PAL score subgroups. Selection criteria: |fold change|> 2, p < 0.05. Red indicates upregulation, blue indicates downregulation. B, C GO and KEGG enrichment analysis of differentially expressed genes. D, E Analysis of somatic mutation features in PAL score subgroups. F Prediction of immune therapy response in PAL score subgroups. G Prediction of small molecule targeted drugs potentially responsive in PAL score subgroups In vitro qRT-PCR validation of prognostic PAL signatures We further explored the expression profiles of PAL prognostic features in vitro cell experiments. As depicted in Fig. [108]9A–F, PCR results indicate significantly decreased expression of [109]AL137782.1 and markedly increased expression of MYOSLID, [110]AC019118.1, ATP2B1-AS1, FLJ21408, and [111]AC092944.1 in the HCT116 cell line. Fig. 9. [112]Fig. 9 [113]Open in a new tab Expression analysis of PAL prognostic features in NCM460 and HCT116 cell lines. A mRNA expression analysis of [114]AL137782.1, B FLJ21408, C ATP2B1-AS1, D MYOSLID, E [115]AC019118.1, and F [116]AC092944.1. Data are presented as mean ± SD (n = 3), *p < 0.05, **p < 0.01, ***p < 0.001 Discussion In this study, we established a prognostic model and verified its reliability by analyzing PAL, which is associated with COAD prognosis. Our results provide new evidence for the role of PANoptosis in COAD. In fact, it has been reported that the key protein of PANoptosis ASC is a pro-apoptotic gene responsible for activating the procaspase-1 pathway in COAD [[117]17]. In addition, COAD prognosis is influenced by the methylation of ASC [[118]18]. The effect of ASC on p53-mediated chemical sensitivity has also been reported in patients with COAD [[119]19]. Together with our results, these evidences support the role of PANoptosis in COAD tumorigenesis. Given the large number of lncRNAs and the limited research available, only two of the six lncRNAs we identified have been reported to potentially influence the mechanisms of COAD. ATP2B1-AS1 has been found to regulate the NF-kappaB signaling pathway [[120]20, [121]21]. Combined with the effect of NF-kappaB signaling pathway on COAD growth, angiogenesis, and tumor metastasis, ATP2B1-AS1 may at least partially affect COAD progression through this pathway [[122]22–[123]24]. MYOSLID, as a competitive endogenous RNA, regulates the expression of MCL-1 [[124]25]. MCL-1 is associated with drug resistance, growth, and apoptosis in colon cancer [[125]26–[126]28]. In addition, MYOSLID regulates the epithelial-mesenchymal transition (EMT) sequence of tumors to promote invasion and metastasis [[127]29]. Therefore, there is a theoretical basis for MYOSLID to influence COAD. Our pathway enrichment experiments showed that Cytoskeleton-related pathway enrichment was the most significant. Cytoskeletal remodeling is one of the important preconditions for tumor cell metastasis [[128]30]. LncRNAs play a significant role in the remodeling of the cytoskeleton in cancer. They can affect cellular functions by directly interacting with actin and its associated proteins, as well as by modulating cytoskeleton-related regulatory pathways, including Rho GTPase and its downstream effector proteins [[129]31]. Multiple lncRNAs participate in malignant behavior of tumors through cytoskeletal remodeling [[130]32]. The cytoskeletal network in colon cancer is also involved in regulating cell polarity, differentiation, proliferation, migration, and invasion [[131]33]. The causality and mechanisms underlying the prognostic differences among various PAL groups and their association with cytoskeleton-related pathways warrant further investigation. In the development of colorectal cancer, a sustained and sustained inflammatory response is essential. Long-term accumulation of colon inflammation significantly increases the risk of colorectal cancer [[132]34]. Myeloid Derived Suppressor Cells (MDSCs) play a key role in the progression of colorectal cancer associated with colitis [[133]35]. MDSCs contribute to tumor progression by enhancing angiogenesis, promoting chronic inflammation, and creating a tumor microenvironment that suppresses immune system activity [[134]36]. MDSC may also promote colitis-associated colorectal cancer progression by promoting the stemness of colon cancer cells [[135]37]. Reducing MDSC can effectively slow tumor progression and produce anti-tumor effects [[136]38]. In addition, the reduction of MDSCs content contributes to the effectiveness of immunotherapy [[137]39]. Our results also showed higher levels of MDSCs in the TME of COAD patients with poor prognosis. As a result, several regimens targeting MDSCs are currently underway in COAD patients [[138]39, [139]40]. In conclusion, we successfully developed the PAL scoring model and preliminarily validated its effectiveness. It is important to note that most of the results in this study were derived from public databases, which may introduce geographical and ethnic biases. Moreover, the bioinformatics analysis primarily involves correlation, with a lack of causal discussion. Therefore, further multi-center, large-scale studies, along with in vitro experiments, are needed to explore the molecular mechanisms more thoroughly. These additional investigations will help confirm the conclusions of this study and enhance our understanding of the role of PAL in COAD. Supplementary Information [140]Supplementary Material 1.^ (9.2KB, xlsx) [141]Supplementary Material 2.^ (9KB, xlsx) Author contributions YW conceived and designed the study. SZ contributed the data collection and data analysis. BZ conceived the original ideas and composed this manuscript. SD and LS designed and completed the experimental section. MX contributed the table and figures of this manuscript. YW and SZ contributed equally to this manuscript. All authors contributed to the article and approved the submitted version. Data availability The data used to support the fndings of this study are available from the corresponding author upon request. Declarations 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. Yangyang Wang and Shihui Zhao contributed equally to this work. References