Abstract Background Colorectal cancer (CRC) is a global health burden because of its high mortalities. It is crucial to discover new biomarkers that can help predict the outcome of patients as well as identify new therapeutic targets to improve treatment options for CRC patients. EGLN1, a protein belonging to the family of prolyl hydroxylase domain-containing proteins, has been associated with the progression of various cancer types. However, its involvement in CRC remains uncertain. Methods We conducted an analysis of EGLN1 expression levels in colorectal cancer (CRC) tissues using publicly available datasets. EGLN1 expression was also evaluated in relation to patient survival. To explore the potential biological functions and signaling pathways associated with EGLN, we performed gene set enrichment analysis (GSEA). We also investigated the immune landscape of EGLN1 in CRC by examining its association with immune infiltration and immune-related markers. Results We consistently observed a significant decrease in EGLN1 level in CRC tumor tissues when compared to normal tissues, indicating its potential role as a tumor suppressor gene specific to CRC. The diagnostic potential of EGLN1 was substantiated by its effective discrimination between normal and tumor tissues in patients with CRC. Functional enrichment analysis further uncovered EGLN1’s involvement in the regulation of crucial biological processes, cellular components, and molecular functions relevant to CRC development and progression. The correlation analysis revealed the potential immunomodulatory role of EGLN1, affecting immune checkpoint molecules, antigen presentation, and immune cell trafficking within the tumor microenvironment. Additionally, high level of EGLN1 exhibited a significant association with improved recurrence-free survival (RFS) and emerged as an independent prognostic factor. Knockdown experiments demonstrated EGLN1’s role in suppressing cell proliferation and migration in CRC. Conclusion In summary, our findings position EGLN1 as a promising multi-faceted biomarker in CRC, with implications for prognosis, immune microenvironment modulation, and potential therapeutic targeting. Keywords: Colorectal cancer, EGLN1, Biomarker, Prognostic, Bioinformatics Introduction Colorectal cancer (CRC) poses a significant challenge to public health. According to a recent study published in CA, there were approximately 153,020 newly diagnosed cases of colorectal cancer (CRC) and about 52,550 deaths for the year 2023 [[30]1]. Despite advancements in CRC diagnosis and treatment, the stage of the disease at the time of diagnosis still has a significant impact on patient prognosis, with higher survival rates observed among early-stage patients compared to those with advanced stages. Therefore, identifying new prognostic biomarkers and therapy targets for CRC holds significant clinical importance. EGLN1, also known as PHD2, is a crucial regulator of cellular response to hypoxia and belongs to the prolyl hydroxylase domain (PHD) enzyme famil [[31]2–[32]4]. Recently, there is growing interest in investigating EGLN1’s potential involvement in cancer, as its dysregulation is observed in several tumor types. However, EGLN1’s prognostic value and probable impact on CRC immune landscape remained largely unexplored. Bioinformatic analyses have been instrumental for elucidating the molecular mechanisms underlying cancer development and identifying potential prognostic biomarkers. In this study, our objective was to comprehensively explore the prognostic significance and immune landscape of EGLN1 in CRC by employing an extensive bioinformatics approach. Specifically, we conducted an analysis of EGLN1 expression levels in CRC tissues using publicly available datasets and examined its correlation with patient prognosis. We conducted gene set enrichment analysis (GSEA) in order to investigate the potential pathways related to EGLN1. Additionally, in order to explore the possible involvement of EGLN1 in modulating immune response, we investigated the correlation of EGLN1 level and immune infiltration in CRC. Our findings may help identify a novel prognostic biomarker and a possible therapy target for CRC. Materials and methods Data collection We obtained RNA sequencing data from publicly available databases, including the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA), along with relevant clinical information for colorectal cancer patients. For the training cohort, we utilized the [33]GSE39582 dataset, while the TCGA-COAD and [34]GSE14333 datasets served as the validation cohort. Differential expression analysis “Limma” package was utilized to assess EGLN1 level among normal and tumor tissues [[35]5]. Additionally, we utilized a paired t-test to assess EGLN1 level among normal and matched tumor tissues. The visualization of the analysis results was carried out utilizing the “ggplot2” package. To assess the diagnostic capability of EGLN1 in differentiating between normal colon tissues and colon tumor tissues, we conducted receiver operating characteristic (ROC) curve analysis [[36]6]. Protein expression analysis Protein Expression Analysis was conducted using cProSite ([37]https://cprosite.ccr.cancer.gov/), an online tool developed and maintained by the Center for Cancer Research (CCR) at the National Cancer Institute (NCI). The data was derived from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) datasets. The statistical significance of differences in protein expression among groups was evaluated using the Mann-Whitney U test. GO and KEGG The patients were categorized into the low EGLN1 group and the high EGLN1 group based on the median. Differential expression analysis was performed using the “limma” package, with a significance threshold of p < 0.05 and an absolute log2 fold change > 0.3 applied to identify the genes that exhibited differential expression between the two groups. The functional annotation of significant differential expression genes (DEGs) was performed using “clusterProfiler” package, which facilitated Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses [[38]7, [39]8]. Statistically significant terms were enriched with p < 0.05. Gene set enrichment analysis (GSEA) We performed gene set enrichment analysis (GSEA) used the “cluster profiler” package [[40]9]. The reference datasets were ‘c2.cp.go.v7.2.symbols.gmt’ and ‘c2.cp.kegg.v7.2.symbols.gmt downloaded from MSigD. The gene sets were ordered according to their correlation with the level of EGLN1. GSEA was conducted by using the enricher function with default settings and p < 0.05 as statistically significant. Survival and prognostic analysis of EGLN1 The optimal threshold for EGLN1 expression was determined using the “surv cutpoint” function available in R package “survminer”. This function identified the cutoff point that maximizes the statistical significance of the survival analysis. Overall survival (OS), recurrence-free survival (RFS) and disease-free survival (DFS) were analyzed using the “survival” package. Survival analysis was performed utilizing Kaplan-Meier (KM) survival curves. The statistical significance of the difference in survival was assessed using the log-rank test. The independent prognostic value of EGLN1 expression was assessed through univariate and multivariate Cox regression analysis. The nomogram served as a graphical tool for prediction of the individual survival probability of CRC patients. The purpose of calibration analysis was to evaluate the agreement between predicted probabilities from the nomogram and the observed outcomes. The C-index was calculated to measure the probability of concordance between predicted and observed survival outcomes [[41]10]. Cell culture The RKO colorectal cancer cell line and HEK293T were obtained from the American Type Culture Collection.The cell was cultured in a controlled environment at 37 °C and 5% CO2 using DMEM supplemented with 10% FBS and 1% penicillin/streptomycin. Sequences targeting EGLN1 were designed using an online shRNA design tool: shEGLN1 #1: 5’-3’ CCGGGACGACCTGATACGCCACTGTCTCGAGACAGTGGCGTATCAGGTCGTCTTTTTG; shEGLN1 #2: 5’-3’ CCGGACGCCACTGTAACGGGAAGCTCTCGAGAGCTTCCCGTTACAGTGGCGTTTTTTG. A scramble sequence, referred to as shNC, was used as a negative control: shNC: 5’-3’ CCGGCGCTGAGTACTTCGAAATGTCCTCGAGGACATTTCGAAGTACTCAGCGTTTTTG. For lentivirus production, HEK293T was simultaneously transfected with the lentivirus and packaging plasmids (psPAX2 and pMD2.G) using Lipo293 Transfection Reagent. Virus supernatant was collected after 48 h and passed through a 0.45 μm filter. RKO cell was then infected with the obtained lentivirus particles. Following a 72-hour incubation, the infected cell was treated with puromycin (2 µg/mL) for a period of 7–10 days, allowing the establishment of stable cell lines. All cell experiments adhered to institutional biosafety guidelines and the International Cell Line Authentication Committee (ICLAC) standards to ensure ethical use of cell lines. Colony formation assay The stable cell line was grown for 10 days at a density of 500 cells per well in six-well plates and then fixed by treating them with a 4% paraformaldehyde solution. After fixation, cells were subjected to 0.1% crystalviolet staining. We captured images of the colonies using a digital camera. Image J was used to count colonies. Scratch wound assay The stable cell line was seeded in 6-well plates and grown until confluence to assess effect of EGLN1 knockdown on cell migration. To create a defined cell-free area, the cell stratum was scraped with a sterile 10-µL pipette tip. PBS was then added to rinse cell debris and better visualize the scratched surface. The cells were then incubated in DMEM without serum for 0, 24, and 48 h. The area of the scratch was measured under a microscope at each time point. Statistical analysis We assessed the characteristics of the tumor microenvironment in each patient using the ESTIMATE algorithm. This algorithm provided insights on tumor microenvironment composition based on gene expression data [[42]11]. To investigate the immune infiltration patterns, the CIBERSORT algorithm was employed [[43]12]. CIBERSORT is a powerful gene expression deconvolution tool that allows for the quantification of different cell types within complex tissues. The LM22 signature matrix was used for the analysis. This matrix contains 547 genes representing 22 immune cell types. Pearson analysis was applied to determine the correlation of EGLN1 with immune-related markers. GraphPad Prism (v. 9.3) and R software (v. 4.3.0) were applied to perform statistical analysis. Wilcoxon rank-sum test or Kruskal-Wallis test was applied for evaluating group differences. Paired t-test was utilized to examine EGLN1 expression level in tumor and matched adjacent normal tissues. Chi-square test was performed to compare the clinical information with EGLN1 level. P-value <0.05 was considered statistically significant. Results Differential expression of EGLN1 in normal and tumor tissues To assess EGLN1 level in colorectal tumors, we conducted an analysis using the [44]GSE39582 dataset, which includes paired samples of normal and tumor tissues. Our analysis found significant higher EGLN1 level in normal tissues than tumor tissues (Fig. [45]1A). Additionally, we consistently observed higher level of EGLN1 in normal tissues compared to paired tumors (Fig. [46]1B). The ROC curve analysis revealed the AUC value was 0.955 (Fig. [47]1C), suggesting that EGLN1 expression levels could effectively differentiate between normal and tumor tissues. To validate our findings, we further analyzed the TCGA-COAD validation dataset. Consistent with the previous analysis (Fig. [48]1D–F), EGLN1 level was higher in normal tissues than in colorectal tumors. Fig. 1. [49]Fig. 1 [50]Open in a new tab Differential level of EGLN1 between normal and tumor tissues. A Boxplot showing EGLN1 differential expression in the [51]GSE39582 dataset. B Boxplot showing EGLN1 differential expression between paired normal and tumor tissues in [52]GSE39582 dataset. C ROC curve analysis of EGLN1 mRNA expression to discriminate normal and tumor tissues in [53]GSE39582 dataset. D Boxplot showing EGLN1 differential expression in the TCGA-COAD dataset. E Boxplot showing EGLN1 differential expression between paired normal and tumor tissues in TCGA-COAD dataset. F ROC curve analysis of EGLN1 mRNA expression to discriminate normal and tumor tissues in TCGA-COAD dataset. G Boxplot showing EGLN1 differential expression at protein level in normal and tumor tissues using cProSite analysis. ****P < 0.0001 These results further supported the conclusion that EGLN1 exhibited reduced expression in colorectal tumor tissues at the RNA level. To evaluate EGLN1 protein expression in CRC, we utilized the cProSite online analysis tool. Our analysis identified a notable elevation in EGLN1 protein levels in normal tissues compared to tumor tissues. (Fig. [54]1G). Overall, our bioinformatics analysis consistently demonstrated that EGLN1 level was markedly increased at both the mRNA and protein levels in normal tissues compared to colorectal tumor tissues. Relationship between EGLN1 and clinical characteristics The low EGLN1 expression group exhibited a higher proportion of stage 3 and 4, increased prevalence of pMMR (mismatch repair), and elevated CIN+ (chromosomal instability). Nevertheless, no statistically significant differences were observed among the remaining clinical characteristics (Fig. [55]2A; Table [56]1). These findings suggested that EGLN1 downregulation may contribute to tumor progression and aggressiveness in CRC. Furthermore, our subgroup analysis demonstrated that EGLN1 expression was particularly low in the CIN + and pMMR groups, suggesting a potential involvement of EGLN1 in modulating the microsatellite instability and mismatch repair (Fig. [57]2B–F). These findings provided significant insights into the involvement of EGLN1 in CRC and its potential clinical implications. Fig. 2. [58]Fig. 2 [59]Open in a new tab Correlation between EGLN1 and clinical characteristics. A Heatmap illustrating the correlation between EGLN1 and clinical features. B–F Boxplots of EGLN1 mRNA expression among different clinical subgroups in colorectal tumors. *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001 Table 1. Clinical characteristics of the sample according to EGLN1 expression status Characteristic EGLN1 high, N = 283^a EGLN1 low, N = 283^a P-value^b Gender Female 123 (43%) 133 (47%) 0.4 Male 160 (57%) 150 (53%) Age <=65 110 (39%) 112 (40%) 0.9 > 65 172 (61%) 171 (60%) Unknown 1 0 Stage Stage 1–2 163 (58%) 134 (48%) 0.018^c Stage 3–4 119 (42%) 146 (52%) Unknown 1 3 mmr.status dMMR 59 (23%) 16 (6.0%) < 0.001^d pMMR 193 (77%) 251 (94%) Unknown 31 16 cin.status – 78 (33%) 32 (14%) < 0.001^d + 162 (68%) 192 (86%) Unknown 43 59 [60]Open in a new tab EGLN1 Egl-9 family hypoxia-inducible factor 1, dMMR different mismatch repair, pMMR proficient mismatch repair, cin chromosomal instability ^an (%) ^bPearson’s Chi-squared test ^cP<0.05 ^dP<0.001 Functional enrichment analysis To explore the functional role of EGLN1 in colorectal tumors, we performed a differential gene expression analysis comparing colorectal tumor samples exhibiting high EGLN1 expression to those displaying low EGLN1 expression. 825 DEGs were identified in the analysis, including 141 downregulated genes and 684 upregulated genes. The heatmap showed the top 20 downregulated and upregulated genes (Fig. [61]3A). The volcano plot displayed a clear separation of the significant DEGs from the non-significant ones (Fig. [62]3B). Fig. 3. [63]Fig. 3 [64]Open in a new tab Differential Gene Expression of EGLN1. A Heatmap of expression profiles about the top 20 significantly downregulated and upregulated genes based on log2FC. B Volcano plot of the top 5 significantly downregulated and upregulated genes based on statistical significance (p-value) The GO-BP analysis of the DEGs revealed significant enrichment in multiple biological processes, including antigen processing and presentation of peptide antigen, immune response, response to oxygen levels and cell-cell adhesion (Fig. [65]4A). The GO-CC analysis identified several significantly cellular components among the DEGs, including the membrane raft, membrane microdomain, secretory granule membrane, and specific granule. (Fig. [66]4B) In the GO-MF analysis, we observed significant enrichment of molecular functions among the DEGs, including chemokine activity, CXCR chemokine receptor binding, MAP kinase tyrosine phosphatase activity and cytokine activity (Fig. [67]4C). Fig. 4. [68]Fig. 4 [69]Open in a new tab Function Enrichment Analysis of EGLN1. A GO-BP analysis according to differential expression genes. B GO-CC analysis according to differential expression genes. C GO-MF analysis according to differential expression genes. D KEGG pathway analysis according to differential expression genes Additionally, the KEGG pathway analysis unveiled several significantly enriched pathways associated with EGLN1 in colorectal tumors. These pathways include HIF-1, B cell receptor, MAPK, TNF, and Chemokine signaling pathways. (Fig. [70]4D). To validate the functional enrichment results, we performed GSEA using a reference dataset. The GSEA results were similar to the findings from the initial GO and KEGG analyses, providing additional support for the functional relevance of EGLN1-associated DEGs in colorectal tumors (Fig. [71]5A, B). Fig. 5. [72]Fig. 5 [73]Open in a new tab Functional analysis of EGLN1 in colorectal tumors using GSEA. A GO enrichment analysis results from GSEA. B KEGG pathway enrichment analysis results from GSEA Correlation analysis of EGLN1 with immune-related markers Our analysis investigating the relationship between EGLN1 expression and immune-related markers in colorectal cancer (CRC) revealed several significant correlations. Specifically, EGLN1 expression was positively correlated with chemokines and their receptors, including CCL18 (r = 0.24, p < 0.001), CCL26 (r = 0.15, p < 0.001), CXCL10 (r = 0.16, p < 0.001), CCR1 (r = 0.20, p < 0.001), CCR2 (r = 0.19, p < 0.001), and CXCR1 (r = 0.17, p < 0.001) (Fig. [74]6A, D). Moreover, EGLN1 expression exhibited positive correlations with major histocompatibility complex (MHC) markers, such as B2M (r = 0.12, p = 0.003), HLA-DMA (r = 0.19, p < 0.001), and HLA-DRA (r = 0.17, p = 0.001) (Fig. [75]6B). This indicates that EGLN1 may play a role in antigen presentation and T cell-mediated immune activation in CRC. Additionally, EGLN1 expression showed positive correlations with immune checkpoint stimulatory factors, such as CD28 (r = 0.15, p < 0.001), CD86 (r = 0.25, p < 0.001), and IL-6 (r = 0.20, p < 0.001) (Fig. [76]6C). These results suggest a potential relationship between increased EGLN1 levels and elevated levels of immune stimulatory molecules, which are essential for immune activation and anti-tumor immune responses. In contrast, EGLN1 expression demonstrated negative correlations with immune checkpoint inhibitory markers, including KIR2DL1 (r = −0.17, p < 0.001) and PDCD1 (r = −0.26, p < 0.001) (Fig. [77]6E). These findings indicate a possible correlation between higher EGLN1 expression and reduced levels of immune checkpoint molecules, known to inhibit antitumor immune responses. Fig. 6. [78]Fig. 6 [79]Open in a new tab Correlation of EGLN1 mRNA with immune-related markers. A Correlation of EGLN1 mRNA expression with chemokine markers. B Correlation of EGLN1 mRNA expression with MHC markers. C Correlation of EGLN1 mRNA expression with immune stimulator markers. D Correlation of EGLN1 mRNA expression with chemokine receptor makers. E Correlation of EGLN1 mRNA expression with immune inhibitor markers. *P < 0.05, **P < 0.01, ***P < 0.001 These findings suggest a potential role for EGLN1 in modulating the trafficking, recruitment, and activation of immune cells within the tumor microenvironment. ESTIMATE score and CIBERSORT analysis The results demonstrated a notable increase in ESTIMATE scores in the EGLN1 high expression group, suggesting a better tumor microenvironment (Fig. [80]7A). In particular, our findings indicated that samples exhibiting high EGLN1 expression had markedly elevated immune and stromal scores, while displaying lower tumor purity when compared to samples with low EGLN1 expression (Fig. [81]7B–D). These analyses showed that elevated EGLN1 level was related to increased immune infiltration and stromal cell content, but decreased tumor cell purity in colorectal cancer. High expression of EGLN1 in CRC was potentially linked to an improved tumor microenvironment. Fig. 7. [82]Fig. 7 [83]Open in a new tab Immunological Analysis of EGLN1 in Colorectal Tumors. A–D Boxplots illustrating the ESTIMATE, immune, stromal, and tumor purity scores between groups with low and high EGLN1 expression. E Boxplots illustrating the fraction of different immune cell types in EGLN1 low and high expression groups by CIBERSORT analysis. F Correlations between immune cell populations and EGLN1 expression based on CIBERSORT analysis. G–I Correlation analysis between EGLN1 expression and Dendritic cells, NK cells, and T cells. *P < 0.05, **P < 0.01, and ****P < 0.0001 The CIBERSORT analysis showed that high EGLN1 level group exhibited higher fractions of T gamma delta cells, NK activated cells, macrophages M0, and neutrophils. Conversely, plasma cells, T regulatory cells, and resting dendritic cells exhibited lower fractions in the high EGLN1 expression group (Fig. [84]7E). The analysis based on CIBERSORT revealed several significant associations between EGLN1 expression and specific immune cell populations. Notably, EGLN1 expression showed positive correlations with Neutrophils (r = 0.28, p < 0.001), M0 Macrophages (r = 0.12, p = 0.004), NK activated cells (r = 0.11, p = 0.006), Dendritic activated cells (r = 0.09, p = 0.04), and T gamma delta cells (r = 0.09, p = 0.03). Conversely, EGLN1 expression exhibited negative correlations with Treg cells (r = −0.26, p = 0.03), NK resting cells (r = −0.10, p = 0.01), Dendritic resting cells (r = −0.13, p = 0.002), CD8 + T cells (r = −0.13, p = 0.001) and plasma cells (r = −0.11, p = 0.01) (Fig. [85]7F–I). Prognostic analysis of EGLN1 The “surv cutpoint” method in R package was utilized to determine optimal cutoff values for EGLN1 expression. The Kaplan-Meier (KM) analysis revealed an association between high EGLN1 level and improved RFS (p < 0.001), while no statistically significant association with OS (p = 0.073) (Fig. [86]8A, B). Another analysis of the [87]GSE14333 dataset, specifically examining DFS, also revealed a statistically significant discrepancy among the two EGLN1 expression groups (p = 0.018) (Fig. [88]8C). High EGLN1 expression linked to better RFS in univariate cox regression analysis (HR = 0.477, 95% CI: 0.281–0.809, p = 0.006) (Fig. [89]8D). Multivariate cox regression analysis adjusting for age, gender and tumor stage confirmed that high expression of EGLN1 was a significant and independent predictor for RFS (HR = 0.583, 95% CI: 0.347–0.988, p = 0.045) (Fig. [90]8E). The nomogram visually depicted the relationship between EGLN1 expression, along with other clinical factors, and the prediction of RFS (Fig. [91]8F). The calibration curve showed good agreement between the predicted and actual rates of RFS at 1 year, 3 years, and 5 years (Fig. [92]8G). The c-index value was found to be 0.669, indicating a moderate predictive ability of nomogram for patient survival. Fig. 8. [93]Fig. 8 [94]Open in a new tab Prognostic value of EGLN1 in CRC. A Kaplan-Meier curve for OS in [95]GSE39582. B Kaplan-Meier curve for RFS in [96]GSE39582. C Kaplan-Meier curve for DFS in [97]GSE14333. D, E The forest plot displays the univariate and multivariate analysis of RFS. D Nomogram predicting RFS based on EGLN1 expression and other clinical characteristics. G Calibration plot illustrating the agreement between observed and predicted RFS probabilities EGLN1 knockdown promotes proliferation and migration in colorectal cancer cells In this study, our objective was to examine the function of EGLN1 in colorectal cancer cell proliferation and migration. We designed two shRNA sequences targeting EGLN1 and generated stable cell lines with knockdown of EGLN1 expression. The number of colonies was markedly higher in EGLN1 knockdown cell than negative control cell (Fig. [98]9A, B). These results suggested that EGLN1 knockdown promoted colorectal cancer cell proliferation. The EGLN1 knockdown cells showed a significantly smaller area between the scratch edges than negative control cells (Fig. [99]9C, D). These results suggested that EGLN1 knockdown promoted colorectal cancer cell migration. Fig. 9. [100]Fig. 9 [101]Open in a new tab Proliferation and migration experiments. A, B Colony formation assays of the stable EGLN1 low expression cell line and the control group. C, D Wound healing assays of the stable EGLN1 low expression cell line and the control group. (magnification, x100). **P < 0.01, ***P < 0.001 and ****P < 0.0001 Discussion EGLN1, a proline hydroxylase, degrades HIF (hypoxia-inducible factor) by catalyzing its hydroxylation under normoxic conditions. This facilitates HIF1α’s recognition by VHL, leading to its ubiquitination and degradation. Conversely, in hypoxic conditions, EGLN1 activity is suppressed, allowing HIF1α to accumulate and activate genes involved in cellular adaptation to low oxygen levels [[102]13–[103]16]. Mutations or abnormal expression of EGLN1 are linked with various diseases including congenital polycythemia [[104]17] cancer [[105]18]ischemic heart disease [[106]19] and pulmonary edema [[107]20]. Interestingly, EGLN1 can act as both a tumor suppressor and an oncogene depending on the context [[108]21]. For instance, it acts as a tumor suppressor by correlating with improved breast cancer survival [[109]22, [110]23], inhibiting hepatocarcinogenesis in mice [[111]24], and suppressing oncogenic pathways via hydroxylation of AKT [[112]25] and SFMBT1 [[113]26]. Conversely, EGLN1 exhibits oncogenic activity by promoting nasopharyngeal carcinoma progression through p53 degradation [[114]27], sustaining clear cell ovarian cancer proliferation [[115]28], and cooperating with c-Myc to drive LSH-mediated lung tumorigenesis [[116]29]. Therapeutically, PHD2 depletion in cancer-associated fibroblasts reduces metastasis [[117]30], while its knockdown in breast cancer cells impedes TGF-β1 processing and growth [[118]31]. In colorectal cancer (CRC), our findings indicate that EGLN1 acts as a tumor suppressor, evidenced by its elevated mRNA and protein levels in normal tissues compared to tumors. The high AUC value from ROC curve analysis suggests EGLN1 could serve as a diagnostic marker for CRC. Low EGLN1 expression correlates with advanced stages, pMMR status, and chromosomal instability, suggesting its role in tumor progression and aggressiveness [[119]32–[120]35]. Functional enrichment analyses of EGLN1-associated differentially expressed genes (DEGs) revealed involvement in immune-related processes, such as antigen processing and presentation, chemokine production, and immune checkpoint modulation. Positive correlations between EGLN1 expression and MHC markers, chemokines, and stimulatory immune checkpoint molecules suggest an immunomodulatory role for EGLN1 in enhancing antitumor immune responses. Additionally, ESTIMATE and CIBERSORT analyses showed that high EGLN1 expression is associated with increased immune infiltration and a favorable immune cell composition, potentially contributing to better patient outcomes. Prognostic analysis demonstrated that high EGLN1 levels significantly correlate with improved recurrence-free survival (RFS) and disease-free survival (DFS) in colorectal cancer (CRC) patients, though no significant difference was observed in overall survival (OS). High EGLN1 levels were identified as an independent prognostic indicator for RFS in Cox regression analysis, suggesting its potential as a prognostic biomarker in CRC. Patients with low EGLN1 expression may require more aggressive treatment strategies to improve survival. To investigate the functional impact of EGLN1 in CRC, we generated stable cell lines with EGLN1 knocked down and performed proliferation and migration assays. Results showed a significant increase in cell proliferation and migration upon EGLN1 knockdown, indicating EGLN1’s crucial role in suppressing these processes. While our study provides novel insights into EGLN1’s role in CRC, further mechanistic experiments are needed to elucidate how EGLN1 regulates immune responses and whether its restoration could serve as a therapeutic strategy. Prospective clinical studies will also be essential to validate its diagnostic and prognostic utility. Conclusion In summary, our findings position EGLN1 as a promising multi-faceted biomarker in CRC, with implications for prognosis, immune microenvironment modulation, and potential therapeutic targeting. Acknowledgements