Abstract Polycystic ovary syndrome (PCOS) is a known risk factor for uterine endometrial cancer (UCEC), but its underlying mechanisms remain unclear. MicroRNAs (miRNAs) could provide insights into these mechanisms and reveal potential therapeutic targets. Differential miRNA expression was analyzed in plasma exosomes from 15 PCOS and 15 control samples. Survival analysis assessed the prognostic value of these miRNAs in UCEC. MiRNA-target gene interaction networks and gene co-expression analyses were used to explore molecular mechanisms. Validation was performed using experimental data from Ishikawa cells treated with six candidate drugs. Among the 15 differentially expressed miRNAs, 12 were up-regulated and 3 were down-regulated in PCOS. Twelve of these miRNAs were associated with UCEC overall survival, with miR-142, miR-424, and miR-331 acting as protective factors, while the remaining 9 miRNAs were identified as risk factors. MiRNA-target network highlighted key genes such as PHF8, LCOR, SFT2D3, E2F1, and ESR1, which were found to be prognostic for patient survival. Further gene expression and co-expression analyses based on miR-424 and miR-330 expression revealed significant alterations in gene expression and cellular processes related to UCEC. Two-sample Mendelian randomization analysis identified potential causal relationships between AURKA gene expression and PCOS or UCEC. Testosterone and estradiol might have adverse roles in UCEC, while drugs like troglitazone, valproic acid, retinoic acid, and progesterone demonstrated various effects on gene expression and cellular processes. Our findings suggest that aberrant miRNA expression, particularly miR-330 and miR-424, may play crucial roles in UCEC progression. The identified miRNAs and candidate drugs may serve as potential therapeutic targets for UCEC, but further research is required to validate and explore their clinical applications. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-01861-4. Keywords: miRNAs, Endometrial cancer, Polycystic ovary syndrome (PCOS), Integrative analysis, Gene expression Introduction Polycystic ovary syndrome (PCOS) is a complex endocrine disorder marked by hyperandrogenism, ovulatory dysfunction, polycystic ovarian morphology, and insulin resistance, with both genetic and environmental factors playing a role [[32]1]. PCOS is associated with various metabolic and reproductive abnormalities, and growing evidence indicates it may also elevate the risk of endometrial cancer (UCEC), one of the most common gynecological malignancies [[33]2]. UCEC is characterized by a wide range of molecular and genetic alterations [[34]3]. Understanding the link between PCOS and UCEC is particularly valuable, as it may shed light on the molecular mechanisms connecting endocrine disorders with cancer development. Recent research has increasingly highlighted the role of microRNAs (miRNAs) as key regulators in cancer biology, influencing various aspects of tumor progression, including growth, metastasis, and resistance to therapy [[35]4]. MiRNAs, small non-coding RNAs that modulate gene expression post-transcriptionally, have emerged as significant biomarkers and potential therapeutic targets in numerous cancers. Plasma exosomal miRNAs are small non-coding RNAs encapsulated within exosomes, nano-vesicles secreted by various cell types [[36]5]. These miRNAs play a pivotal role in intercellular communication, influencing gene expression and cellular functions in distant tissues. They can promote cancer cell invasion, migration, and epithelial-to-mesenchymal transition (EMT) by modulating pathways such as Wnt/β-catenin and STAT3 [[37]6]. Exosomal miRNAs are also important as biomarkers for cancer progression and immune evasion, offering potential targets for therapeutic intervention [[38]7, [39]8]. While some exosomal miRNAs can drive tumor growth, others may suppress it by targeting tumor suppressor genes [[40]9]. Due to their stability in circulation, exosomal miRNAs from various sources, including adipose tissue macrophages, can influence systemic processes such as insulin sensitivity and glucose homeostasis [[41]10]. This study conducted an integrative analysis to identify miRNAs that may link PCOS with UCEC. We found distinct miRNA expression patterns in both PCOS patients and UCEC tissues, with several miRNAs significantly associated with both conditions. Functional validation using cell line experiments explored the effects of candidate drugs targeting miRNA-related genes on gene expression and cellular processes in UCEC. Our findings suggest these miRNAs could serve as potential biomarkers and therapeutic targets, offering insights into the molecular connection between PCOS and UCEC and aiding in personalized diagnosis and treatment strategies. Materials and methods Data acquisition Plasma exosomal miRNA data were obtained from the Gene Expression Omnibus (GEO) database ([42]GSE142819) which included 15 PCOS samples and 15 control samples [[43]11]. 571 UCEC patients from TCGA (The Cancer Genome Atlas) with both gene expression data and miRNA profiles were used for comprehensive analysis [[44]12]. Expression data for Ishikawa cells treated with 6 different chemicals were also downloaded from the NCBI GEO database ([45]GSE69849). For each chemical, three replicates were performed in control and treatment groups [[46]13]. The dataset [47]GSE158317 comprises RNA-Seq data from six ovarian cancer cell line samples (COV318), including three transfected with the miR-330-3p mimic and three with the control [[48]14]. Differential miRNA expression analysis Differential miRNA expression was analyzed using the R software to preprocess and normalize the data. We applied the limma package to identify differentially expressed miRNAs between PCOS and control samples. A volcano plot was generated to visualize significant miRNAs with adjusted P-values < 0.01 and log2 fold change > 1. Survival analysis Survival analysis was conducted using the KM plotter web tool [[49]15]. Kaplan–Meier survival curves were plotted for UCEC patients stratified by high and low expression levels of the identified miRNAs. Statistical significance was determined using the log-rank test [[50]16]. miRNA-target gene interaction network analysis To identify key genes associated with miRNAs, we constructed miRNA-gene interaction networks using the MIENTURNET web tool [[51]17]. Up-regulated and down-regulated miRNAs from PCOS were analyzed separately. Target gene predictions were obtained from databases such as TargetScan and miRDB. The networks were visualized to determine key gene interactions. Survival analysis was conducted on the genes with high connectivity to assess their prognostic value by KM plotter web tool. Differential gene expression and co-expression analysis For UCEC tissues, differential gene expression was assessed based on miR-424 and miR-330 expression levels. The limma package in R was used for differential analysis with adjusted P value < 0.05 and log2 fold change > 0.3. Gene Ontology (GO) and pathway enrichment analyses were performed using the clusterProfiler package [[52]18]. Gene co-expression analysis was conducted using weighted gene co-expression network analysis (WGCNA) to identify gene modules associated with miR-424 and miR-330 [[53]19]. Correlation between module eigengenes and clinical traits was analyzed to identify significant modules [[54]20]. Mendelian randomization analysis To explore potential causal relationships between AURKA gene expression and POCS, UCEC, we performed a two-sample Mendelian randomization (MR) analysis. Summary statistics for AURKA were obtained from eqtl-a-ENSG00000087586, while data for PCOS and UCEC were retrieved from finn-b-E4_POCS and ebi-a-GCST006466 in the MRC IEU OpenGWAS database [[55]21]. Single nucleotide polymorphisms (SNPs) significantly associated with eqtl-a-ENSG00000087586 (p < 5 × 10⁻⁸) were selected as instrumental variables (IVs), ensuring independence (r^2 < 0.001) and absence of linkage disequilibrium. The inverse-variance weighted (IVW) method was applied as the primary MR approach to estimate causal effects, with complementary sensitivity analyses conducted using MR-Egger regression and the weighted median method to account for potential pleiotropy and heterogeneity. The F-statistic was calculated to assess the strength of the IVs. Additionally, heterogeneity was evaluated using Cochran's Q test, and potential horizontal pleiotropy was tested using the intercept from MR-Egger regression. All analyses were performed using the R package TwoSampleMR [[56]22]. Results were reported as odds ratios (OR) or beta coefficients with 95% confidence intervals (CI), representing the effect of eqtl-a-ENSG00000087586 on PCOS and UCEC. Cellular composition analysis Cellular composition profiles of UCEC samples were retrieved using the Ecotyper deconvolution algorithm [[57]23, [58]24]. Heat maps were generated to visualize cellular subtype abundance differences between high and low miR-330 expression samples. The top differentially expressed cell subtypes were identified by t test and their association with patient survival was analyzed using the survminer and survival packages in R. Kaplan–Meier survival curves were plotted for UCEC patients stratified by high and low expression levels of cell subtype abundance. Drug candidate identification Up-regulated and down-regulated genes from miR-330 high samples were used to identify candidate drugs through the enrichr web tool, DSigDB, and literature searches [[59]25, [60]26]. We focused on drugs with known interactions related to hormone signaling and cancer treatment. The Ishikawa cell line experimental data was used for the validation of candidate drugs. Cells were treated with six selected drugs for 6 h. Gene expression changes were identified by limma. Statistical analysis All statistical analyses were performed using R software. P-values < 0.05 were considered statistically significant. Multiple testing corrections were applied where appropriate, using methods such as the Benjamini–Hochberg false discovery rate. Results Differential expression of plasma exosomal miRNAs in polycystic ovary syndrome patients Deep sequencing analysis of the miRNA transcriptome (dataset [61]GSE142819) was performed on plasma samples from 15 control subjects and 15 patients with polycystic ovary syndrome (PCOS). This differential expression analysis identified 15 miRNAs with significant expression changes (Fig. [62]1A). Among them, 12 miRNAs were up-regulated in PCOS patients, including miR-483, miR-206, miR-142, miR-330, miR-3120, miR-203a, miR-1228, miR-1246, miR-3656-p3, miR-4792, miR-193b, and miR-1 (Fig. [63]1B). In contrast, three miRNAs—miR-331, let-7f, and miR-424—were found to be down-regulated in PCOS samples. Fig. 1. [64]Fig. 1 [65]Open in a new tab Differential analysis identified 15 differentially expressed plasma exosomal miRNAs in PCOS patients. A Volcano plot showing differential expression of plasma exosomal miRNAs. B Top ten miRNAs with the most significant fold changes miRNAs as prognostic markers in UCEC tissue PCOS has been associated with an elevated risk of developing UCEC. To explore the prognostic significance of the 15 differentially expressed miRNAs identified in PCOS, we analyzed their association with overall survival in UCEC patients. Notably, 12 of the 15 miRNAs were significantly linked to overall survival, except for miR-1, let-7f, and miR-483. Among these, miR-142 (up-regulated in PCOS), miR-424, and miR-331 (both down-regulated in PCOS) were identified as protective factors. In contrast, the remaining nine miRNAs, all up-regulated in PCOS, were associated with increased risk (Fig. [66]2). These findings suggest that these miRNAs may play a critical role in the heightened risk of developing UCEC in PCOS patients. Fig. 2. [67]Fig. 2 [68]Open in a new tab Survival analysis of tissue miRNAs in UCEC patients grouped by miR expression. Survival curves for UCEC patients stratified by low and high expression levels of miR-142, miR-3120, miR-330, miR-206, miR-3656, miR-4792, miR-1246, miR-1228, miR-193b, miR-203a, miR-424, and miR-331 miRNA and target gene network analysis identifies key genes To elucidate the relationships between miRNAs and their target genes, two miRNA-gene interaction networks were constructed—one for up-regulated miRNAs and the other for down-regulated miRNAs. Through these networks, several key genes were identified. In Network A, genes such as PHF8, LCOR, and SFT2D3 were strongly associated with three specific miRNAs (Fig. [69]3A). In Network B, genes like E2F1, ESR1, C5orf51, VEGFA, and others showed significant connections with various miRNAs (Fig. [70]3B). To validate the relevance of these genes, survival analysis was conducted, revealing that most of the key genes in both networks were prognostic for overall survival in UCEC patients (Fig. [71]3C). These findings suggest that these genes may play critical roles in the progression and prognosis of UCEC. Fig. 3. [72]Fig. 3 [73]Open in a new tab miRNA-gene interaction network reveals key genes. A miRNA-gene interaction network for down-regulated miRNAs. B miRNA-gene interaction network for up-regulated miRNAs. C Survival curves for key genes SFT2D3, E2F1, ESR1, C5orf51, and VEGFA Differential gene expression analysis and co-expression analysis of UCEC stratified by tissue miR-424 expression Given that miR-424 is a protective factor but is down-regulated in PCOS, we explored the differences between UCEC patients with high versus low miR-424 expression. Differential gene expression analysis identified 550 genes (250 up-regulated and 300 down-regulated) in patients with high miR-424 expression compared to those with low expression (Fig. [74]4A). Notably, the most significantly up-regulated genes included SFRP4, ALDH1A2, APOD, CCL21, TWIST2, PAMR1, ECM1, CD248, WNT2, and LTBP4 (Fig. [75]4B). These up-regulated genes were mainly involved in extracellular matrix organization and response to TGF-β signaling (Fig. [76]4C), while the down-regulated genes were associated with ribosome function and mitochondrial gene expression (Fig. [77]4D). Additionally, pathways related to the up-regulated genes were enriched in focal adhesion processes (Fig. [78]4E). Fig. 4. [79]Fig. 4 [80]Open in a new tab Differential gene expression and co-expression analysis based on tissue miR-424 expression in UCEC tissues. A Differential gene expression analysis identified 550 genes in samples with high miR-424 compared to low miR-424. B Boxplots illustrating the top ten significantly changed genes. C–E Gene Ontology (GO) biological processes and pathway enrichment analyses for up-regulated and down-regulated genes. F Heatmap depicting the relationship between four co-expression modules and miR-424. Each cell represents the correlation between the expression of specific modules and distinct clinical parameters, with numerical values in brackets indicating the statistical significance of the correlation Gene co-expression analysis revealed four distinct gene co-expression modules (Fig. [81]4F). Functional enrichment analysis showed that up-regulated module ME1 was related to focal adhesion, while down-regulated module ME2 was linked to the cell cycle. Modules ME3 and ME5 were both associated with ncRNA metabolism. Intriguingly, eight of the top ten significantly up-regulated genes—SFRP4, APOD, CCL21, TWIST2, PAMR1, ECM1, CD248, and WNT2—were all correlated with longer overall survival (Figure S1) and were enriched in the regulation of T cell migration. Further, correlation analysis identified ten genes most significantly associated with miR-424 expression, several of which, including SFRP4, ALDH1A2, and CD248, were linked to the positive regulation of apoptosis. This may help explain why UCEC patients with high miR-424 expression had improved survival rates. Differential gene expression analysis and co-expression analysis of UCEC stratified by tissue miR-330 expression As miR-330 is a risk factor and is up-regulated in PCOS, we explored the differences between UCEC patients with high and low miR-330 expression. Differential gene expression analysis identified 1289 genes (715 up-regulated and 574 down-regulated) in patients with high miR-330 expression compared to those with low expression (Fig. [82]5A). The most significantly altered genes were predominantly up-regulated, including AURKA, ABHD3, NCAPG, YARS, RRM2, AUNIP, and MTFR2 (Fig. [83]5B). These up-regulated genes were primarily involved in nuclear division processes (Fig. [84]5C), while down-regulated genes were associated with the cellular response to TGF-β signaling (Fig. [85]5D). In an independent ovarian cancer cell line dataset [[86]14], we observed that all the most significantly altered genes, except AUNIP, displayed consistent directional changes following treatment with the miR-330 mimic. Fig. 5. [87]Fig. 5 [88]Open in a new tab Differential gene expression and co-expression analysis based on tissue miR-330 expression in UCEC tissues. A Differential gene expression analysis identified 1289 genes in high miR-330 samples compared to low miR-330 samples. B Boxplots showing the top ten significantly changed genes. C–F Gene Ontology (GO) biological processes and pathway enrichment analyses for up-regulated and down-regulated genes Notably, seven of the top ten significantly altered genes were up-regulated, and six of them (AURKA, NCAPG, YARS, RRM2, AUNIP, and MTFR2) were linked to shorter overall survival (Figure S2). These genes were enriched in pathways related to the positive regulation of the cell cycle (Fig. [89]5E). In contrast, the three down-regulated genes—WBP1L, WFS1, and AXIN2—were associated with longer overall survival and were involved in the positive regulation of protein ubiquitination. Pathway enrichment analysis revealed that up-regulated genes were primarily associated with the cell cycle, while down-regulated genes were linked to focal adhesion (Fig. [90]5F). Gene co-expression analysis identified five distinct co-expression modules (Fig. [91]5F). Functional enrichment analysis revealed that the up-regulated modules ME1 and ME3 were involved in cell cycle regulation and immune system processes, while the down-regulated modules ME2, ME4, and ME5 were associated with focal adhesion, TGF-β signaling, and cilium assembly. Interestingly, the top ten genes most strongly correlated with miR-330 expression, identified through correlation analysis, were predominantly involved in the positive regulation of the cell cycle, including AURKA. These findings provide a potential explanation for the poorer survival outcomes in UCEC patients with high miR-330 expression. Validation of the prognostic gene AURKA in UCEC by Mendelian randomization The Mendelian randomization (MR) analysis revealed a significant positive association between AURKA eQTL SNPs and PCOS (Fig. [92]6A). A funnel plot showed a generally symmetric distribution of the genetic variants' estimates, suggesting minimal evidence of directional pleiotropy (Fig. [93]6B). The robustness of these results was further confirmed through sensitivity analyses (Fig. [94]6C). Leave-one-out analysis was performed to assess whether any single SNP disproportionately influenced the overall causal estimate. After sequentially excluding each SNP, the effect estimates remained consistent, demonstrating the stability of the results (Fig. [95]6C). This suggests that no single genetic variant was driving the association between AURKA and PCOS. The similar analysis was also performed for casual relationship between AURKA and UCEC (Fig. [96]6D, [97]F). Taken together, these analyses support robust causal and positive relationships between AURKA and PCOS and UCEC, with minimal evidence of bias from pleiotropy or outlier SNPs. Fig. 6. [98]Fig. 6 [99]Open in a new tab Two-sample Mendelian randomization analysis illustrating the causal effects of AURKA gene expression on PCOS and UCEC. A Scatter plot of effects of cis-eQTL SNPs on AURKA expression and PCOS occurence. B Funnel plot for PCOS. C Leave-one-out sensitivity analysis for PCOS. D Scatter plot of effects of cis-eQTL SNPs on AURKA expression and UCEC occurence. E Funnel plot for UCEC. F Leave-one-out sensitivity analysis for UCEC Comparative analysis reveals differential cell subtypes in UCEC patients stratified by tissue miR-330 expression To investigate the cellular composition differences between UCEC groups stratified by tissue miR-330 expression, we applied the state-of-the-art deconvolution algorithm Ecotyper to the TCGA UCEC samples. The analysis revealed significant heterogeneity in the cellular composition across UCEC samples, as illustrated by the heatmap (Fig. [100]7A). Notably, we identified distinct differences in cellular composition between groups with high and low miR-330 expression. Fig. 7. [101]Fig. 7 [102]Open in a new tab Differences in tissue cellular composition in UCEC patients stratified by miR-330 expression. A Heatmap showing the abundance of cell subtypes in UCEC patients. B Identification of the top 12 differential cell subtypes in high miR-330 samples compared to low miR-330 samples. C Survival curve for samples stratified by abundance of fibroblasts.8. D Survival curve for samples stratified by the abundance of CD4.T.cells.1. In both survival curves, the green line represents high expression and the red line represents low expression The top five significantly altered cell subtypes were epithelial.cells.4, epithelial.cells.2, fibroblasts.8, CD8.T.cells.3, and dendritic.cells.5 (Fig. [103]7B). According to cell state annotation, epithelial.cells.4 is a pro-inflammatory subset, fibroblasts.8 is a pro-migratory-like subset, and CD8.T.cells.3 represents an exhausted T cell state. All three of these subtypes were up-regulated in miR-330 high samples. In contrast, epithelial.cells.2 and dendritic.cells.5, which are normal-enriched subtypes, were down-regulated in miR-330 high samples. Additionally, CD4.T.cells.1, annotated as another exhausted T cell subtype, was up-regulated. Moreover, survival analysis revealed that fibroblasts.8 and CD4.T.cells.1 were both significantly linked to poorer overall survival in UCEC patients (Fig. [104]7C, [105]D). These findings suggest that the aberrant immune cell composition in miR-330 high patients, characterized by increased pro-inflammatory and exhausted cell types, may contribute to the poorer clinical outcomes observed in this group. Identification and validation of UCEC-related drugs based on DEGs from miR-330 high samples We utilized the differentially expressed genes (DEGs) from miR-330 high samples to screen for candidate drugs associated with UCEC. A total of twenty candidate drugs were identified, several of which were hormone-related (Table [106]1). Notably, testosterone was correlated with the up-regulated genes, while two phytoestrogens, enterolactone and coumestrol, were also associated with the up-regulated gene set. Additionally, hormone-related drugs such as calcitriol, retinoic acid, progesterone, and estradiol were identified. These findings highlight the potential involvement of aberrant hormone signaling pathways in the progression of UCEC. Table 1. Identification of candidate drugs based on differential gene expression in high miR-330 samples Name Adjusted P Odds ratio Combined score Up-regulated genes Testosterone 4.1e-104 8.7 2143.7 Enterolactone 5.01e-91 8.9 1925.7 Coumestrol 3.0e-89 6.4 1357.9 Lucanthone 9.3e-89 29.3 6143.0 Dasatinib 1.1e-84 12.8 2572.7 Calcitriol Dihydroxyvitamin D3 1.5e-76 5.5 1007.2 Resveratrol 3.0e-74 5.9 1040.6 Troglitazone 3.3e-59 8.0 1138.5 Etoposide 3.6e-57 316.0 42936.6 Phytoestrogens 5.2e-40 85.1 8194.2 Down-regulated genes Valproic acid 1.6e-24 2.5 160.8 Trichostatin A 2.1e-14 2.2 87.7 Retinoic acid 1.2e-13 2.1 78.4 Progesterone 1.9e-11 2.4 75.9 Vitamin E 1.6e-7 2.2 49.7 Tert-Butyl hydroperoxide 1.6e-7 2.3 50.9 Estradiol 0.0000023 1.7 32.6 Methaneseleninic acid 0.0000023 3.3 63.7 Decitabine 0.0000053 2.0 36.0 Cytarabine 0.0000088 2.7 48.1 [107]Open in a new tab To validate the associations of the candidate drugs, experimental data from the Ishikawa cell line treated with six selected drugs was analyzed. Our findings revealed that testosterone down-regulated estrogen 16-alpha-hydroxylase activity while up-regulating cholesterol metabolism. Estradiol was observed to down-regulate extracellular exosome production while up-regulating developmental processes. These effects suggest potential adverse roles of testosterone and estradiol in UCEC progression. In addition, troglitazone was found to down-regulate artery development while up-regulating cholesterol metabolism. Valproic acid down-regulated nucleoplasm activity and up-regulated cell junction formation. Retinoic acid down-regulated cell migration while up-regulating programmed cell death, indicating its potential as an anti-tumor agent. Lastly, progesterone down-regulated the cell cycle while up-regulating primary metabolism (Fig. [108]8). These experimental data suggest that these drugs could modulate critical biological pathways in UCEC, potentially impacting disease progression and providing insights into therapeutic strategies. Fig. 8. [109]Fig. 8 [110]Open in a new tab Impact of six candidate drugs on gene expression in the Ishikawa cell line of endometrial carcinoma. The bars represent the number of down-regulated and up-regulated genes following 6-h drug treatment. Functional enrichment analysis terms are annotated around the bars, with numbers in parentheses indicating the statistical significance of the enrichment Discussion In this study, we performed an integrative analysis to investigate the role of miRNAs in linking PCOS with UCEC, identifying several miRNAs as potential biomarkers and therapeutic targets. Our analysis revealed 15 differentially expressed miRNAs in PCOS, many of which were also associated with UCEC prognosis. Notably, up-regulated miRNAs were generally linked to poorer survival outcomes, while down-regulated miRNAs were protective. For instance, miR-331 and miR-424 are involved in processes like metabolic reprogramming and hormonal regulation, which may drive cancer progression by influencing pathways such as estrogen signaling and cellular proliferation [[111]27, [112]28]. MiR-1246 was identified as a signature that could discriminate high-grade serous ovarian cancer patients with high accuracy [[113]29]. The ceRNA networks MALAT1/miR-203a/TGFβR2 and [114]AK128202/miR-483-5p/GOT2 are implicated in regulating proliferation, angiogenesis, and insulin resistance in PCOS. As such, they have been proposed as potential biomarkers and therapeutic targets for PCOS [[115]30]. MiR-330, known for promoting tumorigenesis in various cancers, was highly expressed in PCOS and linked to adverse outcomes in UCEC, suggesting its role in connecting the two conditions [[116]31]. MiR-330 is highly expressed in both cancer tissues and serum of breast cancer patients, and it can enhance axillary lymph node metastasis, a significant factor influencing breast cancer prognosis [[117]32]. MiR-330 is significantly up-regulated in gestational diabetes mellitus (GDM) patients and is a potential biomarker for glucose homeostasis and pregnancy outcomes in GDM [[118]33]. These results underscore the potential of these miRNAs as prognostic markers for UCEC. Gene co-expression analysis highlighted key interactions between miR-330 and genes like SFT2D3 and E2F1, while miR-424 was associated with PHF8 and LCOR. Interestingly, we used miWalk to confirm the associations of these miR-target gene pairs (data not shown) [[119]34]. Functional enrichment indicated that up-regulated genes in miR-330-high samples were involved in nuclear division and cell cycle regulation, while TGF-β signaling was more active in miR-424-high samples. AURKA is a vital gene in the cell cycle and TGF-β signaling, which may induce an inflammatory response during PCOS progression [[120]35]. These results underscore the importance of miRNA regulation in processes like cell division, extracellular matrix organization, and TGF-β signaling, all critical in cancer progression [[121]36, [122]37]. We also found significant immune cell composition changes linked to miR-330 expression, with pro-inflammatory and exhausted immune subsets being up-regulated in high miR-330 samples, indicating an unfavorable immune microenvironment in UCEC. These findings suggest that miR-330 may shape the immune landscape in a way that worsens prognosis. Our drug screening identified hormone-related agents such as testosterone, estradiol, and progesterone as influential in miR-330-related pathways. Interestingly, estradiol and progesterone have been identified as potential therapeutic drugs for PCOS [[123]38]. Testosterone was found to up-regulate cholesterol metabolism while down-regulating estrogen 16-alpha-hydroxylase activity, reflecting its complex role in UCEC. The up-regulation of cholesterol metabolism may indicate a metabolic reprogramming in UCEC cells that could be detrimental to disease progression. Progesterone, a hormone frequently used in UCEC treatment, was observed to down-regulate the cell cycle while up-regulating primary metabolism. These effects are consistent with progesterone’s known ability to inhibit proliferation and promote metabolic stability in cancer cells [[124]39]. These findings align with previous research indicating that hormonal imbalances play a crucial role in UCEC development and progression [[125]40–[126]43]. In addition to hormone-related agents, other drugs identified in our screening also showed significant potential in modulating key pathways in UCEC. Troglitazone, known for its impact on metabolic pathways [[127]13], was found to down-regulate artery development while up-regulating cholesterol metabolism. This dual action suggests that troglitazone may influence both tumor vascularization and lipid management, processes critical to tumor growth and survival. Valproic acid, an HDAC inhibitor [[128]43], demonstrated the ability to down-regulate nucleoplasm components, potentially suppressing gene expression or cell division, while simultaneously up-regulating cell junction integrity, which may enhance cellular adhesion and slow cancer cell dissemination. Retinoic acid, a modulator of cellular differentiation [[129]42], down-regulated cell migration while promoting programmed cell death, positioning it as a promising candidate for UCEC therapy by inhibiting tumor spread and encouraging apoptosis. Together, these findings highlight the diverse mechanisms by which non-hormonal drugs can influence UCEC progression and offer potential therapeutic avenues beyond hormonal regulation. In conclusion, this study, for the first time, provides valuable insights into the molecular link between PCOS and UCEC by identifying key miRNAs and their associated pathways. The role of miRNAs in immune modulation, hormonal signaling, and metabolic reprogramming offers new opportunities for therapeutic intervention. Future research should focus on validating these findings and developing targeted therapies to improve outcomes for patients with both PCOS and UCEC. Limitations This study has several limitations. First, although the sample size in both PCOS and UCEC datasets was sufficient for exploratory analysis, larger cohorts may enhance the robustness of the findings. Second, the identified miRNA-gene interactions and potential therapeutic targets are based on computational predictions; therefore, further experimental validation, including qRT-PCR, Western blotting, miRNA knockdown models, and immune cell models in both clinical and in vitro settings, will be important to confirm their biological significance and elucidate the underlying mechanisms. Finally, our drug screening results are promising, but preclinical studies will be needed to substantiate their potential therapeutic applications. Supplementary Information [130]12672_2025_1861_MOESM1_ESM.docx^ (320.6KB, docx) Additional file 1: Figure S1 Up-regulated genesSFRP4, APOD, CCL21, TWIST2, PAMR1, ECM1, CD248, and WNT2were all correlated with longer overall survival in UCEC samples with high miR-424 expression. Figure S2 Up-regulated genes AURKA, NCAPG, YARS, RRM2, AUNIP, and MTFR2 were linked to shorter overall survival. Down-regulated genesWBP1L, WFS1, and AXIN2were associated with longer overall survival in UCEC samples with high miR-330 expression. Author contributions J.Y. Conceived and designed the study. X.L. and L.W. performed the experiments. P.L. performed the data statistical analysis. L.Y. prepared the final figures. X.L. wrote the paper. All authors read and approved the final manuscript. Funding The authors received no specific funding for this work. Data availability The datasets used in this study are available from the GEO and TCGA databases. Declarations Ethics approval and consent to participate Not applicable. 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. Xuedan Lai and Ling Wu contributed equally to this work. References