Abstract Breast cancer is a leading malignancy in women, with mortality disparities between developed and underdeveloped regions. Accumulating evidence suggests that the competitive endogenous RNA (ceRNA) regulatory networks play paramount roles in various human cancers. However, the complexity and behavior characteristics of the ceRNA network in breast cancer progression have not been fully elucidated. The expression profiles of three RNAs (long non-coding RNAs [lncRNAs], microRNAs [miRNAs], and mRNAs) were extracted from breast cancer and adjacent samples were sourced from the TCGA database. The SLC26A4-AS1- hsa-miR-19a-3p-NTRK2 ceRNA network related to the prognosis of breast cancer was obtained by performing bioinformatics analysis. Importantly, we identified the SLC26A4-AS1/NTRK2 axis within the ceRNA network through correlation analysis and found it to be a potential prognostic model in clinical outcomes based on Cox regression analysis. Moreover, methylation analysis suggests that the aberrant downregulation of the SLC26A4-AS1/NTRK2 axis might be attributed to hypermethylation at specific sites. Immune infiltration analysis indicates that the SLC26A4-AS1/NTRK2 axis may have implications for the alteration of the tumor immune microenvironment and the emergence and progression of immune evasion in breast cancer. Finally, we validated the expression of SLC26A4-AS1-hsa-miR-19a-3p-NTRK2 in breast cancer cell lines. In summary, the present study posits that the SLC26A4-AS1/NTRK2 axis, based on the ceRNA network, could be a novel and significant prognostic factor associated with breast cancer diagnosis and outcomes. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-02080-7. Keywords: Breast cancer; LncRNA; MiRNA; MRNA; CeRNA network; Prognosis,; Methylation; Immune infiltration Introduction Breast cancer (BC) stands as the predominant malignant tumor in women, securing a spot within the top three most diagnosed cancers globally. According to GLOBOCAN 2022, BC remains the most frequently diagnosed cancer in women, with an estimated 2.3 million new cases and 685,000 deaths globally in 2022, accounting for 11.6% of all new cancer cases and 6.9% of all cancer-related deaths. While developed regions have observed a notable decline in breast cancer-related deaths, primarily due to early detection and effective systemic treatments, breast cancer remains a leading cause of cancer-related deaths in underdeveloped regions, where late-stage diagnoses and limited access to treatment are significant challenges [[34]1]. Therefore, the persistent advancement in breast cancer diagnosis and therapy becomes paramount to curbing its mortality. Thus, continuous updates in breast cancer diagnosis and treatment are essential for reducing mortality rates in breast cancer. The public availability of comprehensive cancer genomic datasets, notably The Cancer Genome Atlas (TCGA), offers an abundance of clinical and molecular insights for diverse cancer types, serving as a pivotal resource in pinpointing potential tumor biomarkers. The burgeoning use of high-throughput RNA sequencing (RNA-seq) has catalyzed the creation of enhanced tools, recognized for their non-invasiveness, reproducibility, sensitivity, and precision, benefiting early screenings, diagnoses, patient evaluations, and continual monitoring [[35]2]. Interestingly, while roughly 75% of the human genome can generate functional transcripts, a mere 2% encode proteins, directing the spotlight to non-coding RNAs (ncRNAs) which encompass lncRNAs, miRNAs, circular RNAs, and pseudogenes [[36]3]. Despite mRNA research being relatively mature, investigations on lncRNAs, miRNAs, and pseudotranscripts remain nascent, with vast uncharted territories awaiting exploration [[37]4]. Defined as non-coding RNA variants spanning over 200 nucleotides with little to no protein-coding capabilities, lncRNAs engage in modulating numerous cellular activities through a myriad of channels, encompassing transcriptional governance via interactions with DNA [[38]5], RNA [[39]6], and proteins [[40]7], and more. Their influence spans epigenetic modifications, protein/RNA stability, translation, post-translational modifications, and even direct interactions with signaling receptors [[41]8]. The pivotal roles of lncRNAs in cancer biology, especially their frequent anomalies in cancers [[42]9], including BC, underline their clinical significance. Some lncRNAs have been associated with cancer relapses, metastasis [[43]10], and post-radiotherapy complications [[44]11]. Therefore, investigating the role of lncRNAs in the prognosis and diagnosis of BC holds significant clinical importance. On another note, microRNAs (miRNAs) are diminutive, intrinsic, and highly conserved ncRNAs, orchestrating nearly 30% of gene expressions post-transcriptionally. Typically, they hinder mRNA translation and stability, thereby governing genes pivotal in cellular functions like inflammation, cell cycle, stress reactions, differentiation, apoptosis, and migration [[45]12]. Moreover, the intricate play of miRNA-mediated regulation leans heavily on an assortment of RNA-binding proteins, granting miRNAs the capacity to influence tumor onset and development. Within the cytoplasmic realm, Salmena et al. [[46]13] proposed the intriguing competing endogenous RNA (ceRNA) hypothesis, which portrays a dynamic RNA transcript communication network. By competing for shared miRNA sequences, these transcripts adjust one another's expressions. This theory underscores pivotal discoveries concerning the interplay between coding and non-coding RNAs mediated by miRNA. Earlier research illustrated that lincRNA-RoR (long intergenic noncoding RNA regulator of reprogramming) could bolster the invasive prowess of triple-negative breast cancer via the miR-145/ARF6 route by destabilizing E-cadherin positioning [[47]14], a key epithelial adhesion molecule that maintains intercellular junctions and tissue integrity. The loss of E-cadherin destabilizes cell–cell adhesion, promoting cytoskeletal rearrangement and enhancing cellular motility, processes central to epithelial-mesenchymal transition (EMT). EMT not only drives breast tumor invasion and metastasis but also contributes to chemoresistance and radio-resistance by enabling tumor cells to acquire mesenchymal-like traits and evade therapeutic interventions, underscoring the therapeutic potential of targeting EMT-regulating factors like lncRNAs and miRNAs [[48]15]. Furthermore, the PI3K-Akt signaling pathway, a key regulator of proliferation, survival, and migration, is frequently dysregulated in breast cancer and has been associated with resistance to various therapies, making it a crucial target for potential interventions [[49]16]. In addition, Chou et al. [[50]17] highlighted that the lncRNA MALAT1 escalates breast cancer cell migration and intrusion by competing for miR-1, catalyzing the surge of cell division control protein 42 (CDC42). Contrarily, Kim et al. [[51]18] discerned an inverse relation between MALAT1 concentrations and breast cancer advancement, with MALAT1 acting more as a metastasis suppressor. It acts as a suppressor of tumor metastasis rather than a promoter. Therefore, unraveling the complex interactions of specific lncRNA-miRNA-mRNA axes through high level bioinformatics and network analysis is crucial for deciphering these pathways and identifying specific targets for breast cancer diagnosis and treatment. In this study, we hypothesize that specific ceRNA axes, such as SLC26A4-AS1-hsa-miR-19a-3p-NTRK2, play critical roles in the progression of breast cancer by influencing gene expression through epigenetic modifications and modulating the tumor immune microenvironment. To test this hypothesis, we constructed a tri-regulatory lncRNA-miRNA-mRNA ceRNA network specific to BC and identified SLC26A4-AS1-hsa-miR-19a-3p-NTRK2 as a key axis associated with breast cancer prognosis. We further explored the functional mechanisms of this axis through methylation assessments, immune infiltration analysis, and pathway enrichment studies, integrating bioinformatics approaches and experimental validation using the GEO database and breast cancer cell lines. Our findings reveal that the ceRNA-based SLC26A4-AS1/NTRK2 axis may serve as a novel and significant prognostic factor in breast cancer, providing potential therapeutic targets for future research. Materials and methods Data preparation and preprocessing We obtained mRNA sequencing (mRNA-seq) data for 1113 cases of BC and 113 adjacent non-cancerous samples, as well as miRNA sequencing (miRNA-seq) data for 1103 cases of BC and 104 adjacent non-cancerous samples, from the TCGA database ([52]https://portal.gdc.cancer.gov/). Survival information for the TCGA-BC dataset was sourced from UCSC Xena ([53]http://xena.ucsc.edu/). We divided the publicly available data into cancerous and adjacent non-cancerous groups. We conducted differential analysis on the raw count matrix of the selected public data using the DESeq2 package, following the standard procedure. Furthermore, we applied the Variance Stabilizing Transformations (VST) method provided by the DESeq2 package to normalize the raw count matrix. For protein expression analysis, we utilized the UALCAN portal ([54]http://ualcan.path.uab.edu/analysis-prot.html). In addition, we obtained gene expression profiles from the Gene Expression Omnibus (GEO, [55]https://www.ncbi.nlm.nih.gov/GEO/) database, specifically [56]GSE45827 (Platform: [57]GPL570) and [58]GSE12790 (Platform: [59]GPL570), to further validate our findings. Screening of differentially expressed genes (DEGs) Differential expression lncRNAs (DElncRNAs) and differential expression mRNAs (DEmRNAs) were identified with thresholds of |logFC|> 1 and p.adj < 0.05. The differential expression miRNAs (DEmiRNAs was identified with thresholds of |logFC|> 0.5 and p.adj < 0.05. Volcano plots for visualizing DEGs were generated using the R package ‘‘EnhancedVolcano’’. Heatmap clustering of DEGs was constructed using the R package ‘‘pheatmap’’. Establishment of the ceRNA network in BC In accordance with the hypothesis that lncRNAs act as natural sponges in the cytoplasm, competitively regulating mRNA expression indirectly through miRNAs, the construction steps for the ceRNA network are as follows: (1) Utilizing miRNet ([60]https://www.mirnet.ca) to predict potential miRNAs targeted by DElncRNAs and lncRNA-miRNA interactions; (2) Using TargetScan ([61]http://www.targetscan.org) and miRDB ([62]http://www.mirdb.org) to predict target genes by overlapping the predicted miRNAs with DEmiRNAs; (3) Employing the R package ‘‘VennDiagram’’ to facilitate the intersection between target genes and DEmRNAs. selecting the resulting mRNA intersection for subsequent analysis; (4) Integrating the lncRNA-miRNA pairs with the miRNA-mRNA pairs to construct the lncRNA-miRNA-mRNA triple-regulation network. The generated network was visualized using Cytoscape software (version 3.9.1). LncRNA sequences were obtained from LNCipedia ([63]https://lncipedia.org), and lncRNA cellular localization was determined using the LncLocator database ([64]http://www.csbio.sjtu.edu.cn/bioinf/lncLocator/). Functional enrichment analysis We conducted functional enrichment analysis on DEmRNAs in the lncRNA-miRNA-mRNA triple-regulation network using the R software package ‘‘Goplot’’ and ‘‘clusterProfiler’’. Significance thresholds for GO terms and KEGG pathways were set at p.adj < 0.05. Additionally, we obtained a set of 20 NTRK2-binding proteins based on the STRING database ([65]https://cn.string-db.org) and supported by experimental evidence. We analyzed the correlation among these genes pairwise in BC, then visualized the correlation results using the ‘‘circlize’’ package and performed GO enrichment analysis (Molecular fuction; MF, Cellular component; CC, Biological process; BP) and KEGG pathway analysis. Visualization of the results was carried out using the R package ‘‘ggplot2’’. Survival analysis and construction of survival-related ceRNA in BC Survival status and time in BC patients were sourced from TCGA clinical dataset, filtered for female gender and no history of neoadjuvant treatment. Overall survival (OS) was defined as the time from diagnosis to death from any cause. Disease-specific survival (DSS) was defined as the time from diagnosis to death specifically caused by breast cancer. Progression-free interval (PFI) was defined as the time from diagnosis to disease progression or recurrence. Patients without these events at the end of follow-up were censored. We conducted batch survival regression analysis on the ceRNA network using the Cox proportional hazards model in the “survival” package in R to candidate lncRNAs, miRNAs, and mRNAs associated with OS, DSS, and PFI. Kaplan–Meier survival curves were generated to visualize survival trends, and hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated to assess the impact of gene expression on survival outcomes. Based on the expression levels of candidate genes, we performed time-dependent receiver operating characteristic curve (ROC) analysis using timeROC package to assess the predictive capability of biomarkers, calculating the area under the curve (AUC). The results were visualized using ‘‘ggplot2’’. When the value of a molecule (risk factor) trends towards promoting event occurrence, the AUC for that molecule is > 0.5. Larger areas (AUC value closer to 1) indicate better prognostic efficacy. Conversely, when a molecule (protective factor) trends in the opposite direction of event occurrence, the AUC for that molecule is < 0.5. Smaller areas (AUC values closer to 0) indicate better prognostic efficacy. Methylation and expression analysis of NTRK2 DNA methyltransferases (DNMT1, DNMT3A and DNMT3B) are critical players in epigenetic regulation, with roles ranging from the maintenance of existing DNA methylation patterns to the establishment of new ones. Their impact on gene expression and epigenetic memory makes them important subjects of study in the field of molecular biology and genetics. First, we investigated the expression levels of three DNA methyltransferases in NTRK2-high and NTRK2-low groups using the TCGA database. Subsequently, we assessed the methylation levels of NTRK2 between BC and adjacent normal tissues using the DiseaseMeth 3.0 version ([66]http://bio-bigdata.hrbmu.edu.cn/diseasemeth/) and UALCAN. Furthermore, we analyzed the methylation levels of NTRK2 CpG sites in BC using MethSurv ([67]https://biit.cs.ut.ee/methsurv/) and examined the correlation between these CpG sites and the survival of breast cancer patients. Finally, we employed SMART ([68]http://www.bioinfo-zs.com/smartapp/) to investigate the methylation values of different CpG sites of NTRK2 in BC and normal tissues and analyzed the relationship between these CpG sites and NTRK2 expression. Immune infiltration levels and expression analysis of NTRK2 We employed the ssGSEA algorithm provided in R package GSVA to calculate the immune infiltration status in BC using markers for 24 immune cell types as provided by Bindea et al. [[69]19]. Subsequently, we analyzed the correlation between NTRK2 and these immune cells and visualized using ‘‘ggplot2’’. Additionally, based on the median expression level of NTRK2 (4.435, derived from the log2-transformed expression values) in BC samples, we stratified the samples into two groups: high-NTRK2 expression group (≥ 4.435) and low-NTRK2 expression group (< 4.435). This stratification allowed us to analyze the differences in immune infiltration between these two groups. Furthermore, we used the TIDE ([70]http://tide.dfci.harvard.edu/query/) to evaluate the relationship between NTRK2 expression and CTL and assess the relationship between NTRK2 and T-cell exclusion score, T-cell dysfuction score, response to immune checkpoint blockade (ICB) therapy, and gene knockout phenotype in CRISPR screens. Cell culture Human MCF 10A (American Type Culture Collection, USA) and MDA-MB-468 cells (Cell Bank of the Chinese Academy of Sciences, China) were propagated in Dulbecco’s modified Eagle’s medium (DMEM; Corning) supplemented with 10% fetal bovine serum. Cells were cultured in standard T-25 or T-75 culture flasks (Corning) and maintained in an incubator at 37 °C with 5% CO₂ and saturated humidity. The medium was replaced every 2–3 days, and cells were subcultured at 70–80% confluence. Cell morphology and viability were monitored under an inverted phase-contrast microscope before each experiment, and cells were regularly tested for Mycoplasma contamination using a Mycoplasma Detection Kit (Biyuntian, C0303S). Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) Cells were washed twice with PBS, collected by centrifugation, and lysed with Trizol Reagent (Shanghai Pufei Biotech Co., Ltd., Shanghai, China) to extract total RNA, including the miRNA fraction, following the manufacturer's instructions. RNA purity and concentration were determined using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific), and integrity was verified by agarose gel electrophoresis. For reverse transcription, 2 μg of total RNA was used with M-MLV reverse transcriptase (Promega) at 43.5 °C for 60 min, followed by enzyme inactivation at 73.5 °C for 3 min. qPCR reactions were carried out using LightCycler 480 SYBR Green (Roche) on a LightCycler 480 II (Roche). The reaction mix (12 μL) included 6.0 μL SYBR Green premix, 0.5 μL each of forward and reverse primers (5 μM), 1.0 μL cDNA, and 4.0 μL RNase-free water. qPCR cycling conditions were as follows: initial denaturation at 95 °C for 5 min, followed by 40 cycles of denaturation at 95 °C for 15 s, annealing at 60 °C for 20 s, and extension at 72 °C for 20 s. Melt curve analysis confirmed amplification specificity. Primers for qPCR were purchased from Ribobio and Shanghai Sangon Biotech. The specific primers for SLC26A4-AS1 were 5′-TTTCTGCTCCTTGTTGCTT-3′ (forward) and 5′-TGCCTCCTGTTGTTACTCA-3′ (reverse); for hsa-miR-19a-3p, they were ssD809230917 (forward; Ribobio), SSD089261711 (reverse; Ribobio), and ssD809230225 (RT; Ribobio); for NTRK2, they were 5′-TCACCTTGACTTGTCTGAACTG-3′ (forward) and 5′-CTGGACTGGATTTAGCCTCTT-3' (reverse). The primers for ACTB were 5′-GCGTGACATTAAGGAGAAGC-3′ (forward) and 5′-CCACGTCACACTTCATGATGG-3′ (reverse); for U6, they were ssD0904071008 (forward; Ribobio), ssD0904071006 (reverse; Ribobio), and ssD0904071007 (RT; Ribobio). The ΔΔCt method was used for data analysis, with ACTB and U6 serving as internal controls. Western blotting The cells were lysed in RIPA buffer (Biyuntian, P0013), and total proteins were quantified using the BCA Protein Assay Kit (Biyuntian, P0010S). Equal amounts of protein (20–30 µg) were separated on a 10% denaturing polyacrylamide gel and transferred onto a PVDF membrane (Millipore) at 4 °C, 200 mA for 120 min. Membranes were blocked with 5% non-fat milk in TBST for 1 h at room temperature and incubated overnight at 4 °C with primary antibodies: anti-NTRK2 (Proteintech, 13,129-1-AP, rabbit, 1:3000) and anti-β-actin (Santa Cruz, sc-69879, mouse, 1:13,000). After washing, membranes were incubated with HRP-conjugated secondary antibodies (CST, #7074 for anti-rabbit IgG; #7076 for anti-mouse IgG, 1:8000) for 1.5 h at room temperature. Protein bands were visualized using the LumiGLO® Reagent and Peroxide Kit (CST, #7003), and images were captured on X-ray films. NTRK2 (90–95 kDa) and β-actin (43 kDa) served as the target and loading control, respectively. Statistical analysis In this study, all data analysis and visualization were performed using GraphPad Prism software (version 9.0.0) and R software (version 4.2.1; [71]https://www.r-project.org/) and the corresponding packages. A p-value of less than 0.05 was considered statistically significant. Results Identification of differentially expressed genes (DEGs) between BC and adjacent normal samples. A total of 1113 BC samples and 113 adjacent normal samples for identifying DElncRNAs and DEmRNAs, and 1103 BC samples and 104 adjacent normal samples for identifying DEmiRNAs. In BC samples, we identified 4177 DElncRNAs, of which 64.3% (2686) were upregulated and 35.7% (1491) were downregulated. Similarly, 5065 DEmRNAs were identified, with 62.1% (3146) upregulated and 37.9% (1919) downregulated. For DEmiRNAs, we identified 118 differentially expressed miRNAs, including 62.7% (74) upregulated and 37.3% (44) downregulated, using stringent criteria (|log2 FC|> 0.5 and p.adj < 0.01). Volcano plots and heatmaps provided a visual representation of the distribution of DEGs between BC and adjacent normal samples, as well as the expression of 20 significant variable genes (Fig. [72]1). Fig. 1. [73]Fig. 1 [74]Open in a new tab Visualization of Differentially Expressed RNAs and Corresponding Heatmaps. A The volcano plot highlights the DElncRNAs. Accompanying the plot, a heatmap is presented, showcasing the expression patterns of the top 20 most variable DElncRNAs. B Similarly, a volcano plot for the DEmiRNAs is displayed. Adjacent to it, a heatmap elucidates the expression profiles of the top 20 most variable DEmiRNAs. C A volcano plot captures the DEmRNAs, with an adjoining heatmap revealing the expression trends of the top 20 most variable DEmRNAs Construction of the lncRNA-miRNA-mRNA triple regulatory network DelncRNAs were subjected to miRNet database, which predicted 631 potential miRNAs targeting them. Among these, 14.1% (89) overlapped with the identified DEmiRNAs. These 89 miRNAs were further used to predict mRNA targets through TargetScan and miRDB databases, yielding 2309 mRNAs by intersecting with the 5065 DEmRNAs, accounting for 45.6% of the total DEmRNAs. (Fig. [75]2A). GO and KEGG enrichment analyses were conducted on these mRNAs, with a significant threshold of corrected p < 0.05. GO enrichment analysis revealed enrichment in development and structural organization GO terms in Biological Processes (BP), including urogenital system development (GO: 0001655), external encapsulating structure organization (GO: 0045229), nuclear division (GO: 0000280), renal system development (GO: 0072001), and extracellular matrix organization (GO: 0030198). Cellular Component (CC) included providing structural organization and cellular compartmentalization enriched GO terms, mainly associated with collagen-containing extracellular matrix (GO: 0062023), synaptic membrane (GO: 0097060), basement membrane (GO: 0005604), condensed chromosome (GO: 0000793), and chromosome, centromeric region (GO: 0000775). Molecular Functions (MF) were enriched in fundamental biological processes and molecular interactions GO terms, with notable terms such as extracellular matrix structural constituent (GO: 0005201), transmembrane receptor protein kinase activity (GO: 0019199), transmembrane receptor protein tyrosine kinase activity (GO: 0004714), glycosaminoglycan binding (GO: 0005539), and DNA-binding transcription activator activity (GO: 0001216). Additionally, fundamental cellular processes and signaling KEGG pathways were identified, including Cell cycle including Cell cycle (hsa04110), Focal adhesion (hsa04510), PI3K-Akt signaling pathway (hsa04151), ECM-receptor interaction (hsa04512), and Axon guidance (hsa04360) (Fig. [76]2B). Finally, a triple regulatory network comprising 192 lncRNAs, 89 miRNAs, and 2309 mRNAs (Supplementary Fig. 1) was constructed using Cytoscape. Moreover, we conducted batch survival regression analysis on the network, and a triple regulatory network associated with BC prognosis-related genes was generated (Fig. [77]2C). Fig. 2. [78]Fig. 2 [79]Open in a new tab Construction of the lncRNA-miRNA-mRNA Network and Its Functional Enrichment Analysis. A Venn diagrams display the overlap between DEmiRNAs and the miRNAs targeted by DElncRNAs, as well as the intersection between DEmRNAs and the mRNAs targeted by shared miRNAs from the previous overlap. B Visualization of the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses for the intersecting DEmRNAs and their predicted mRNA counterparts. C A highlighted subset of genes within the ceRNA co-regulatory network that have associations with breast cancer prognosis Identification of a prognostic ceRNA network in BC To identified key ceRNA with significant prognosis value in BC, we analyzed the status and prognostic value of hub triple regulatory network RNAs associated with prognosis in BC. Our results showed 6 upregulated and 3 downregulated lncRNAs, 6 upregulated and 4 downregulated miRNAs, 33 upregulated mRNAs, and 18 downregulated mRNAs in BC compared to adjacent normal breast tissues (Fig. [80]3). Survival analysis revealed the high expression of 1 lncRNA (LINC01588), 2 miRNAs (hsa-miR-9-5p and hsa-miR-1307-3p), and 7 mRNAs (FAM83D, SDC1, ANLN, CCNE1, CENPA, LRP8, and CENPN) was associated with the worst OS, DSS, and PFI prognosis, while high expression of 5 lncRNAs (MAPT-IT1, LINC01224, SLC26A4-AS1, LINC02568, and DIO3OS) and 14 mRNAs (CCL19, ABCB1, NEFH, IL2RB, KRT17, FCRLA, GREB1L, NTRK2, NR3C2, ETNPPL, ABAT, ZNF703, DMD, and CCL5) was associated with best prognosis (Fig. [81]4). Fig. 3. [82]Fig. 3 [83]Open in a new tab Distribution of Expression Patterns for 73 Prognostically-Associated Hub-RNAs Derived from the Triple Regulatory Network in the TCGA BRCA Dataset. A Expression patterns of 9 pivotal long non-coding RNAs (hub-lncRNAs) in BRCA samples. B Expression distribution of 10 key microRNAs (hub-miRNAs) within BRCA samples. C Expression trends of 54 central messenger RNAs (hub-mRNAs) observed in BRCA samples Fig. 4. [84]Fig. 4 [85]Open in a new tab Prognostic Efficacy of the 73 Hub-RNAs Derived from the Triple Regulatory Networks. A Forest plots displaying the overall survival, disease-specific survival, and progression-free survival of lncRNAs. B Forest plots presenting the survival outcomes for miRNAs, encompassing overall survival, disease-specific survival, and progression-free survival. C Forest plots illustrating the survival profiles of mRNAs with regards to overall survival, disease-specific survival, and progression-free survival Considering the upregulated LINC001588 interacted with the upregulated hsa-miR-9-5p, and SLC26A4-AS1 was mainly localized in the cytoplasm, whereas LINC001588 was mainly distributed in the cytosol (Fig. [86]5A). Thus, SLC26A4-AS1 may act as a ceRNA in BC, sequestering hsa-miR-19a-3p to regulated mRNAs expression. Furthermore, expression correlation analysis showed that ADRB1, ABCB1, MDFIC, NTRK2, NR3C2, GABRA2, and SOCS3 were positively correlated with SLC26A4-AS1 and negatively correlated with hsa-miR-19a-3p expression (Fig. [87]5B), among which, we observed a significant decrease in NTRK2 total protein levels in BC, consistent with mRNA expression levels using data from CPTAC (Fig. [88]5C). Therefore, we constructed the SLC26A4-AS1-hsa-miR-19a-3p-NTRK2 ceRNA network. Target prediction of SLC26A4-AS1 and NTRK2 3' UTRs paired with miR-19a-3p was conducted using the starBase version 2.0 database ([89]http://starbase.sysu.edu.cn) (Fig. [90]5D). We also observed a significant downregulation of NTRK2 in BC patients and breast cancer cell lines in the GEO databases [91]GSE45827 and [92]GSE12790, further validating NTRK2 expression. Thus, we selected the SLC26A4-AS1/NTRK2 axis in the ceRNA network as a potential prognostic model for further analysis (Fig. [93]5E). Fig. 5. [94]Fig. 5 [95]Open in a new tab Development and Comprehensive Analysis of the ceRNA Network. A Inference of the cellular localization for SLC26A4-AS1 and LINC01588. B Correlational analysis involving SLC26A4-AS1, hsa-miR-19a-3p, and associated mRNAs in BRCA. C Quantitative representation of the protein expression of genes interacting with SLC26A4-AS1 in normal and primary tumor tissues of BRCA; other mRNAs were undetected in the Breast dataset. D Conceptual model delineating the ceRNA network, accompanied by predicted binding site indications. Overexpressed genes are marked in red, and underexpressed genes in blue. E Empirical validation of NTRK2 expression within breast tissues and breast cancer cell lines, utilizing the GEO database.onstruction and integrated analysis of the ceRNA network. Notations: *p < 0.05, **p < 0.01, ***p < 0.001 Clinical relevance of the SLC26A4-AS1- hsa-miR-19a-3p-NTRK2 ceRNA network in BC patients. SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 are candidate genes for constructing BC specific prognostic biomarkers. Grouping gene expression by median, K-M survival analysis showed that patients with high expression of SLC26A4-AS1 (Fig. [96]6A) and NTRK2 (Fig. [97]6B) had higher OS than those with low expression, while patients with high expression of hsa-miR-19a-3p had lower OS than those with low expression (Fig. [98]6C). It is noteworthy that SLC26A4-AS1, NTRK2, and hsa-miR-19a-3p do not significantly predict prognosis in the early stages (I-II) of BC but have a strong predictive capability for the prognosis in late-stage (III-IV; Supplementary Fig. 2). Time-dependent ROC curve analysis confirmed that SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 have significant prognostic value (Fig. [99]6). We also analyzed correlations between SLC26A4-AS1- hsa-miR-19a-3p-NTRK2 ceRNA network and the pathological stage of BC patients, and we found that low NTRK2 expression was significantly correlated with earlier tumor stage (I-II), while expression levels of SLC26A4-AS1 and hsa-miR-19a-3p were not (Supplementary Fig. 2A–C). Fig. 6. [100]Fig. 6 [101]Open in a new tab Prognostic Assessment of Predictive RNAs in BRCA. A Comprehensive analysis encompassing expression profiling, Kaplan–Meier survival estimation, and time-dependent receiver operating characteristic (ROC) curve evaluation for SLC26A4-AS1. B Parallel evaluations for NTRK2. C Analogous analyses for hsa-miR-19a-3p Furthermore, we explored the correlation between the expression levels of SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 with clinical factors. Our study showed that the expression levels of SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 were significantly associated with PR status and ER status (p < 0.001), as shown in Supplementary Table 1. However, no significant correlation was observed between the expression levels of these genes with other clinical factors, including age, menopause status, pathologic T stage, pathologic N stage, pathologic M stage, pathologic stage, HER2 status, and anatomic neoplasm subdivisions (p > 0.05). In addition, in the univariate cox regression analysis models of SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2, some prognostic factors (Age, Pathologic T stage, Pathologic N stage, Pathologic M stage, Pathologic stage, Menopause status) were closely associated with OS in BC patients in the TCGA cohort (p < 0.05). Importantly, in the multivariate Cox regression analysis, aberrant expression of SLC26A4-AS1 remained associated with OS in breast cancer patients, while aberrant expression of hsa-miR-19a-3p and NTRK2 was not related to poor prognosis (Supplementary Fig. 3A-B). Therefore, SLC26A4-AS1 may serve as an independent prognostic factor in BC patients. The methylation status of NTRK2 in BC To further elucidate the mechanism underlying the downregulation of NTRK2 in BC tissues, we explored the correlation between NTRK2 expression levels and its methylation status through various methods. Firstly, differential expression analysis of three DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) between breast cancer NTRK2-high and NTRk2-low groups showed a significant increase in DNMT3B expression in the NTRK2-low group compared to the NTRK2-high group, although DNMT1 and DNMT3A exhibited no significant differences (Fig. [102]7A). Secondly, UALCAN analysis revealed that the methylation levels of NTRK2 were higher in breast cancer tissues than in normal breast tissues (Fig. [103]7B). Similarly, analysis using DiseaseMeth 2.0 showed that the methylation levels of NTRK2 were significantly higher in BC tissues compared to normal tissues (Fig. [104]7C). Fig. 7. [105]Fig. 7 [106]Open in a new tab Methylation analysis of NTRK2. A Comparative expression of three DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) between high-NTRK2 and low-NTRK2 expressing BRCA samples. B Analysis of NTRK2 methylation patterns in BRCA via the UALCAN platform. C Examination of NTRK2 methylation status in BRCA utilizing the DiseaseMeth version 2.0 tool. D Discrepancies in methylation intensities of CpGs within NTRK2 between BRCA and normal tissue samples. Correlational investigation between methylation values across multiple CpG sites and NTRK2 expression magnitude in BRCA. E Heatmap representation highlighting the clustering of CpG methylation degrees within NTRK2 in BRCA, processed through the Methsurv database. Methylation levels are denoted continuously, with a spectrum ranging from blue (fully unmethylated, 0) to red (fully methylated, 1). Each row signifies a specific CpG, whereas columns represent individual patients. F Survival implications depicted via a forest plot, focused on diverse CpG sites within NTRK2 in BRCA. Notations: ns indicates no significance; *p < 0.05, **p < 0.01, ***p < 0.001 Interestingly, through analysis using the SMART App, we found that several CpGs within the NTRK2 gene exhibited higher methylation levels in BC samples compared to normal sample, including cg01009697, cg03628748, and cg08470639. Furthermore, the methylation (beta-value) of these CpGs was negatively correlated with NTRK2 expression. In addition, when compared to normal tissues, cg13620631, cg14273545, and cg13654445 exhibited lower methylation levels in BC, but except for cg13620631, cg14273545, and cg13654445 methylation was positively correlated with NTRK2 expression (Fig. [107]7D). Subsequently, we utilized MethSurv to present the methylation status of multiple CpGs within the NTRK2 gene in BC in the form of a heatmap (Fig. [108]7E). It is noteworthy that high methylation of CpGs cg13620631, cg13698224, cg14273545, and cg01009697 was associated with higher overall survival rates in TCGA breast cancer patients, while high methylation of cg01292475 and cg13965062 associated with lower overall survival rates in breast cancer patients (Fig. [109]7F). Correlation between NTRK2 expression and immune infiltration in BC Tumor-infiltration cells are an important component of the tumor microenvironment and are closely associated with the occurrence, development, or metastasis of cancer. We assessed the potential correlation between NTRK2 expression and immune infiltration levels in breast cancer. The results showed that NTRK2 was negatively correlated with the infiltration levels of Th2 cells, Treg, aDC, and NK CD56dim cells, but positively correlated with CD8 T cells, DC, Eosinophils, iDC, Mast cells, Neutrophils, NK CD56bright cells, NK cells, pDC, T cells, T helper cells, Tcm, Tem, Th17 cells, and TReg (Fig. [110]8A). Additionally, breast cancer tissues with low NTRK2 expression showed lower infiltration levels of B cells, CD8 T cells, DC, Eosinophils, iDC, Mast cells, Neutrophils, NK CD56bright cells, NK cells, pDC, T cells, T helper cells, Tcm, Tem, and TFH, while aDC, TReg, and Th2 cells showed higher infiltration levels (Fig. [111]8B). Fig. 8. [112]Fig. 8 [113]Open in a new tab Immune Landscape Pertaining to NTRK2 in BRCA. A Association of NTRK2 expression with the extent of immune cell infiltration in BRCA tissues. B Discrepancies in immune cell infiltration between NTRK2 high-expressing and low-expressing BRCA cohorts. C Elucidation of the interrelation between NTRK2 expression and cytotoxic T-lymphocyte (CTL) infiltration. D Exploration of the interplay between NTRK2 expression and various parameters: T-cell exclusion metric, T-cell dysfunction index, responsiveness to immune checkpoint blockade (ICB) interventions, and the phenotypic manifestation in CRISPR-based gene ablation studies. Notations: ns indicates no significance; *p < 0.05, **p < 0.01, ***p < 0.001 There are two main mechanisms of immune evasion in cancer: (1) inducing T-cell dysfunction in tumors with high cytotoxic T lymphocyte (CTL) infiltration, and (2) preventing T-cell infiltration in tumors with low CTL levels. To predict whether NTRK2 was involved in BC immune evasion, we explored the correlation between NTRK2 and CTL using the TIDE database. The results showed that NTRK2 was negatively correlated with CTL infiltration in BC (Fig. [114]8C). Figure [115]8D showed that NTRK2 was associated with a positive T-cell dysfunction score in the Metabric dataset. Additionally, NTRK2 expression levels were higher in tumor-associated M2 macrophages (TAM M2) and lower in CAF FAP, cell types known to promote T-cell exclusion. These findings suggest that NTRK2 may contribute to immune dysregulation and facilitate immune evasion in BC patients through mechanisms involving T-cell exclusion. Exploring NTRK2-associated proteins and pathways in BC To further screen NTRK2-targeted binding proteins and investigate the molecular mechanism of NTRK2 in BC, we retrieved 40 NTRK2 binding proteins (medium confidence) from the STRING database. Figure [116]9A displays the protein–protein interaction network. KEGG pathway enrichment analysis revealed that neurotrophin signaling pathway, Ras signaling pathway, and PI3K-Akt signaling pathway are the most significantly involved in NTRK2's impact on BC (Fig. [117]9B). Furthermore, the expression level of NTRK2 remained unaffected in breast cancer cell lines treated with a PI3K-Akt signaling pathway inhibitor (Fig. [118]9C), suggesting that NTRK2 may be upstream of the PI3K-Akt signaling pathway. Additionally, GO enrichment analysis related to NTRK2, including biological processes (Fig. [119]9D), cellular components (Fig. [120]9E), and molecular functions (Fig. [121]9F), mainly enriched in signaling, synaptic function, and kinase activity regulation. Finally, the results of SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 expression were validated in breast cancer cell MDA-MB-468 and breast epithelial cell line MCF-10A (Fig. [122]9G). However, the observed changes in NTRK2 protein in breast cancer cell were not parallel changes in NTRK2 RNA levels (Fig. [123]9H). Fig. 9. [124]Fig. 9 [125]Open in a new tab Exploration of NTRK2-Associated Gene Enrichment and Expression Validation. A Correlational scrutiny of 20 experimentally ascertained NTRK2-binding proteins in BRCA samples. B KEGG pathway exploration anchored on NTRK2-associated genes. C Evaluative analysis of the influence of PI3K-Akt pathway inhibitors on NTRK2 expression in JIMT1 and HCC1954 breast cancer cell lines. D–F GO enrichment analysis for NTRK2-associated genes distributed across Biological Processes D, Cellular Component E, and Molecular Function (F). G Quantitative assessment of mRNA expression levels for SLC26A-AS1, hsa-miR-19a-3p, and NTRK2 employing RT-qPCR. H The protein expression magnitude of NTRK2 ascertained through Western blotting analysis. Notations: ns indicates no significance; *p < 0.05, **p < 0.01, ***p < 0.001 Discussion Breast cancer is one of the three most common cancers worldwide. While early-stage breast cancer is considered curable, advancements in treatment have shifted the focus towards reducing both overtreatment and undertreatment. Identifying reliable biomarkers is crucial for developing new therapeutic targets and improving patient prognosis. Increasing evidence has revealed that non-coding RNAs (ncRNAs) play pivotal roles in breast cancer progression. However, the intricate regulatory interplay between different types of ncRNAs remains poorly understood. In this study, we established a lncRNA-miRNA-mRNA tri-regulatory network in BC, identifying 192 lncRNAs, 89 miRNAs, and 2,309 mRNAs. Enrichment analysis highlighted the network’s focus on key processes such as "extracellular matrix organization" and "extracellular matrix receptor interaction." This suggests that these ncRNA interactions may influence tumor progression by modulating the extracellular matrix, contributing to either tumorigenic or tumor-suppressive effects. Subsequent prognostic analysis identified a core ceRNA network of 9 lncRNAs, 10 miRNAs, and 54 mRNAs, among which the SLC26A4-AS1-hsa-miR-19a-3p-NTRK2 axis emerged as significantly associated with prognosis in BC. Expression profiling and survival analyses further confirmed that low expression of this axis correlates with poor clinical outcomes. The roles of SLC26A4-AS1 and NTRK2 in cancer have been reported previously. Yuan et al. [[126]20] found that lncRNA SLC26A4-AS1 was significantly downregulated in thyroid cancer and lymph node metastasis specimens, and low expression was linked to poor prognosis. They also demonstrated that SLC26A4-AS1 inhibits the MRE11/RAS50/NBS1 complex-mediated DNA repair signal, thereby suppressing thyroid cancer metastasis by disrupting DDX5. In addition, SLC26A4-AS1 has been shown to promote autophagy and mitigate thyroid papillary carcinoma progression by recruiting ETS1 to enhance ITPR1 expression [[127]21]. Similarly, Li et al. [[128]22] identified SLC26A4-AS1 downregulation in gastric cancer, where it was also significantly associated with poor prognosis. Consistent with these findings in other cancers [[129]23, [130]24], our study demonstrates that low expression of SLC26A4-AS1 in BC is linked to poor prognosis, and for the first time, we validated this in the breast cancer cell line MDA-MB-468. Currently, research on hsa-miR-19a-3p mainly focuses on non-cancer domains. For instance, reduced expression of hsa-miR-19a-3p was found in premenopausal women with genital arousal disorder [[131]25] and in patients with spinal cord injury-induced neuropathic pain [[132]26], while its expression was aberrantly increased in patients with vitiligo [[133]27]. We are the first to identify and validate the abnormally high expression of hsa-miR-19a-3p in BC, which is associated with poor prognosis in BC patients. This suggests that the aberrant expression of hsa-miR-19a-3p is related to the onset and development of human cancers. The Tropomyosin Receptor Kinases (TRKs) were first identified in 1986 when Neurotrophic Receptor Tyrosine Kinase 1 (NTRK1) was discovered to be a part of an oncogenic fusion gene in colorectal cancer. Shortly thereafter, NTRK2 and NTRK3 were also identified. NTRK2, also known as Trk-β, encodes the TRKB protein, which binds to neurotrophins and regulates neuronal development and function [[134]28]. It has been reported that NTRK fusions are common in some rare types of tumors, such as secretory carcinoma of adult breasts and salivary glands, as well as infantile fibrosarcoma (IFS) in pediatrics, with reported fusion positivity rates often exceeding 75% [[135]29, [136]30]. Furthermore, drugs targeting NTRK fusions are currently in clinical trials, with some, like TRK inhibitors larotrectinib and entrectinib, being approved as tumor-agnostic treatments for solid tumors harboring NTRK fusions [[137]31]. In addition, Gomez et al. [[138]32] found that overexpression of NTRK2 in non-small cell lung cancer is associated with in vitro proliferation and invasion. Hu et al. [[139]33] concluded that NTRK2, as a target oncogene of miR-22, is involved in the in vitro development of gastric cancer. However, current research on NTRK2 in breast cancer is limited, and its expression in breast cancer differs from its expression in other tumors. A recent study showed that NTRK2 is abnormally downregulated in breast cancer cell lines MCF-7 and MDA-MB-231 [[140]34]. Similarly, based on the TCGA database, our results show that the expression of SLC26A4-AS1 and NTRK2 in BC is significantly lower than in normal tissue. Survival analysis revealed that low expression of SLC26A4-AS1 and NTRK2 is associated with poor prognosis. In contrast, the expression of hsa-miR-19a-3p is upregulated, and its high expression level indicates a poorer OS prognosis for BC. Additionally, we validated the mRNA levels of NTRK2 in the [141]GSE12790 and [142]GSE45827 datasets. DNA methylation is the most extensively studied epigenetic modification, playing a pivotal role in promoting embryonic development, genomic imprinting, and X-chromosome inactivation, among other vital biological processes. Aberrations in DNA methylation can lead to alterations in the cellular microenvironment, impacting gene expression patterns, ultimately giving rise to various pathological conditions, including carcinogenesis [[143]35]. In recent years, emerging non-invasive DNA methylation biomarkers have shown significant clinical value in predicting cancer prognosis and drug responses [[144]36]. Therefore, we consulted various online sources to explore DNA methylation patterns that could elucidate the aberrant expression of NTRK2 in BC. We discovered that, compared to adjacent normal samples, the methylation level of NTRK2 in BC samples is elevated. This is consistent with the downregulation of NTRK2 in BC observed when combined with DNA methyltransferase analysis (DNMT1, DNMT3A, and DMNT3B; Supplementary Fig. 4). We also noticed that the low expression group of NTRK2 significantly co-occurred with high expression of DNMT3B, providing insights into the downregulation of NTRK2 in BC. Additionally, compared to normal tissue, we identified specific methylation sites of NTRK2 in BC samples that displayed higher methylation levels and showed a negative correlation with NTRK2 expression. In contrast, some sites with low methylation in BC positively correlated with NTRK2 expression and negatively correlated with the prognosis of BC patients. Lastly, we observed that high methylation in the TSS1500 region (g01009697), body region (cg13698224 and cg14273545), and the 3'UTR region (cg13620631) of NTRK2 resulted in a higher overall survival rate for TCGA breast cancer patients. Conversely, elevated methylation in the TSS200 region (cg01292475) and 5'UTR (cg13965062) resulted in a lower overall survival rate. These findings suggest that aberrant methylation may be associated with BC prognosis, and more evidence is needed to validate the potential role of NTRK2 DNA methylation in the onset of BC tumors. The tumor microenvironment (TME) is a principal regulator of tumor initiation, progression, and therapeutic responses. The TME comprises stromal cells (including myoepithelial cells, fibroblasts, and vascular endothelial cells), immune cells (such as T cells, B cells, macrophages, and natural killer cells), and the extracellular matrix. Most cancer pathobiology research has predominantly focused on changes and drivers within cancer cells, with relatively less attention paid to the surrounding milieu. Compared to other cancers like melanoma or lung cancer, the breast cancer microenvironment exhibits significant cellular and spatial heterogeneity [[145]37]. Tumor-infiltrating lymphocytes (TILs) are immune cells that migrate to malignant lesions as part of the immune response promoting tumor elimination. They encompass various immune cells, primarily T cells (CD8 + cytotoxic T cells and CD4 + helper or regulatory T cells (Tregs)), as well as B cells and NK cells. A systematic review encompassing 15 studies involving nearly 14,000 breast cancer patients revealed the highest level of TIL infiltration in triple-negative breast cancer (TNBC), followed by HER2-positive breast cancer, with the luminal subgroups exhibiting the lowest TIL infiltration [[146]38]. Moreover, in a retrospective analysis of over 5,000 female breast cancer samples, low Treg abundance correlated with pCR post-neoadjuvant chemotherapy in TNBC women but was unrelated to ER-positive/HER2-negative disease [[147]39]. These findings indicate the presence of immune infiltrating cells varies considerably within breast cancer subtypes and between different subtypes. Dendritic cells (DCs) play a pivotal role in antigen presentation during immune surveillance and orchestrating anti-tumor immune responses. DCs isolated from breast cancer tumors [[148]40] and peripheral blood [[149]41] of breast cancer patients have been shown to exhibit impaired maturation and reduced T cell stimulatory activity. Tregs in the TME exhibit immunosuppressive actions, promoting the growth and spread of malignant tumors. Through secreting inhibitory cytokines, Tregs can foster intra-tumoral T cell exhaustion by modulating the expression of inhibitory receptors in CD8 + TILs and exhaustion-associated transcriptomic features [[150]42]. In our study, we found low NTRK2 expression correlates with reduced TIL and DC infiltration, but a higher degree of TReg infiltration. Tumors with high cytotoxic T lymphocyte (CTL) infiltration can induce T cell dysfunction, leading to tumor immune evasion. Our results also show NTRK2 negatively correlates with CTL infiltration in BC, with elevated expression levels in tumor-associated M2 macrophages (TAM M2) and reduced expression in CAF FAP. These findings suggest aberrant NTRK2 expression relates to immune dysregulation in BC patients and may promote immune evasion via T cell exclusion. Consequently, the role and functionality of NTRK2 in immunotherapy for BC warrant further investigation. To further explore the biological functions of NTRK2, we conducted GO and KEGG enrichment analyses for NTRK2 and its associated proteins. The enriched pathway related to NTRK2 is the PI3K-Akt signaling pathway, and analysis of the [151]GSE69189 dataset suggests that NTRK2 might be positioned upstream in the PI3K-Akt signaling pathway, consistent with earlier research results [[152]43]. GO enrichment analyses associated with NTRK2 (including BP, CC, and MF) were primarily enriched in signaling, synaptic function, and regulation of kinase activity. Lastly, we confirmed the mRNA expression of SLC26A4-AS1, hsa-miR-19a-3p, and NTRK2 in breast cancer cell line MDA-MB-468 and breast epithelial cell line MCF-10A. However, we did not observe any differential expression of NTRK2 protein between breast cancer cell lines and normal breast cells. The inconsistency between NTRK2 mRNA and protein levels suggests that reduced NTRK2 RNA levels might be common, but they may not correlate with the actual total protein expression or the expression of NTRK2 in BC might involve complex transcriptional and post-transcriptional regulatory mechanisms involving non-coding RNA, an issue that deserves further investigation. Although we have constructed the SLC26A4-AS1/NTRK2 axis based on the ceRNA mechanism, which appears to be a potential prognostic biomarker for clinical application, certain limitations must be acknowledged. First, the binding affinities of lncRNAs, miRNAs, and mRNAs retrieved from databases require further experimental validation. Secondly, the function and mechanism of the SLC26A4-AS1/NTRK2 axis in the initiation and progression of BC need to be further explored through in vitro and in vivo studies. In conclusion, we have established a low-expression ceRNA network related to BC prognosis (SLC26A4-AS1-hsa-miR-19a-3p-NTRK2), which offers deeper insights into the correlations among lncRNA-miRNA-mRNA. Additionally, we found that the SLC26A4-AS1/NTRK2 axis based on the ceRNA mechanism might be a novel and critical prognostic factor involved in BC. This prognostic model contributes to our understanding of the pathogenic mechanism of BC. Supplementary Information [153]Supplementaty material 1.^ (21.8MB, docx) [154]Supplementaty material 2.^ (22.3KB, docx) Acknowledgements