Abstract Background Long non-coding RNAs (lncRNAs) play crucial roles in the progression of breast cancer (BC). The lncRNA small nuclear RNA host gene 16 (SNHG16), represents a lncRNA associated with tumor development. This study focuses on SNHG16 and its regulatory role in BC progression, validated through microarray analysis and in vitro experiments. Method The GEO datasets [41]GSE65194 and [42]GSE61304 were used to identify differentially expressed lncRNAs, [43]GSE41922 and [44]GSE45666 for differentially expressed miRNAs, and [45]GSE29431 and [46]GSE42568 for differentially expressed mRNAs. The TCGA database was used to validate the genes identified in the previous analyses. The regulatory pathways of SNHG16 were identified through differential gene analysis, target gene prediction, functional enrichment analysis, and protein-protein interaction network analysis. RT-qPCR, Western blot, CCK-8, cell migration, and cell knockdown experiments were performed for validation. Results Our study found that elevated expression levels of SNHG16 were associated with BC cell proliferation and poor prognosis. According to an in vitro knockdown assay, SNHG16 silencing inhibits the progression of BC. SNHG16 was found to positively regulate AURKA levels as a competitive sponge for hsa-let-7b-5p. Conclusion The activity of the lncRNA SNHG16 can be triggered via the hsa-let-7b-5p/AURKA axes. These findings shed light on the novel molecular mechanisms underlying BC progression. Keywords: Breast cancer, SNHG16, ceRNA network, Proliferation, Migration Highlights * • SNHG16 is significantly differentially expressed in breast cancer through TCGA and GEO databases. * • The lncRNA SNHG16 is involved in the ceRNA regulatory network. * • qRT-PCR showed that SNHG16 was highly expressed in breast cancer cell lines. * • Knockdown of SNHG16 decreased the proliferation and metastasis of breast cancer cells. 1. Introduction Breast cancer (BC) is a malignant tumor originating in the epithelial tissue of the breast, commonly causing female mortality and increasing incidence among young women [[47]1,[48]2]. Advances in treatment have significantly reduced mortality rates, but the primary cause of death remains metastasis rather than the primary tumor [[49]3]. Research shows that over 90 % of breast cancer-related deaths are due to metastasis, highlighting its critical role in mortality [[50]4,[51]5]. Therefore, identifying molecular biomarkers for early detection and understanding metastasis mechanisms are crucial for effective treatment. The complex interplay of multiple genes, factors, and molecules drives the development of breast cancer. Long non-coding RNAs (lncRNAs) are found in the nucleus or cytoplasm, they regulate gene expression via competing endogenous RNA (ceRNA) mechanisms. Dysregulated expression of lncRNAs contributes to disease progression through ceRNA regulatory networks [[52][6], [53][7], [54][8]]. The ceRNA hypothesis proposes that lncRNAs and mRNAs can competitively bind microRNAs (miRNAs) through shared microRNA response elements (MREs) [[55][9], [56][10], [57][11], [58][12]]. This interaction reduces miRNA-mediated suppression of mRNA, influencing biological processes such as proliferation, migration, and apoptosis in tumor cells [[59]13,[60]14]. Recent studies have shown that lncRNA SNHG16 negatively regulates miR-218-5p expression in pancreatic cancer, thereby inhibiting cancer cell invasion and metastasis [[61]15,[62]16]. In breast cancer, SNHG16 similarly acts as a molecular sponge, regulating downstream target genes and thus affecting migration [[63]17]. Downregulation of has-let-7b in ovarian cancer promotes oncogenic gene expression, enhancing cell proliferation and migration [[64]18]. Elevated miR-196a-5p levels in triple-negative breast cancer cells promote proliferation and migration, which decrease upon inhibition of upstream genes [[65]19]. Research has validated that AURKA impacts cell proliferation in hepatocellular, gastric, and breast cancer cells [[66][20], [67][21], [68][22], [69][23]]. However, whether SNHG16 acts as a ceRNA to regulate downstream AURKA expression in breast cancer remains unexplored. This study uses the TCGA and GEO databases to identify differentially expressed RNAs related to breast cancer prognosis, constructing a ceRNA expression network. Experimental validation of this ceRNA network's impact on breast cancer proliferation and invasion provides new biomarkers and theoretical foundations for understanding disease progression. 2. Materials and methods 2.1. Microarray analysis The Cancer Genome Atlas (TCGA) is the largest sequencing database dedicated to breast cancer research [[70]24]. We screened 943 eligible breast cancer tissue samples and 95 adjacent normal tissue samples, merged the data from both using Perl scripts, and compiled mRNA expression profiles for 60,488 Ensembl genes and miRNA expression profiles of 1882 genes. Genes were annotated using the Homo sapiens genome information file (Homo_sapiens.GRCh38.105.chr.gtf), distinguishing between lncRNAs and mRNAs. Meanwhile, the Gene Expression Omnibus (GEO) is the most comprehensive repository of microarray data worldwide [[71]25]. For our study, we selected the breast cancer gene expression microarrays [72]GSE65194 and [73]GSE61304 to identify differentially expressed lncRNAs; [74]GSE41922 and [75]GSE45666 for miRNAs; and [76]GSE29431 and [77]GSE42568 for mRNAs. Detailed dataset information is provided in [78]Table S1. 2.2. Identification of differentially expressed genes (DEGs) We conducted RNA sequencing analysis using the limma package in R to group, deduplicate, and analyze RNA samples. DEGs were defined based on log[2] (fold change) > 2 or log[2] (fold change)<-2 (log[2]FC>|2|) and with P < 0.05. Visualization of DEGs was achieved using ggplot2 for volcano plots and pheatmap for heatmaps. Upregulated genes were marked in red, while downregulated genes were marked in green. We performed an intersection analysis of DEG results across various databases and gene chips. 2.3. Functional enrichment analysis of DEGs Functional characterization and enrichment analysis of DEGs were conducted using Gene Ontology (GO) terms, categorizing biological processes (BP), molecular functions (MF), and cellular components (CC). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis identified significant pathways enriched with DEGs compared to the whole genome background. Visualization of enrichment results was performed using ggplot2 in R (4.2.0). 2.4. Protein-protein interaction (PPI) network analysis of DEGs PPI networks illustrate interactions between proteins, providing insights into downstream protein expression regulation. DEGs were analyzed using the STRING database, and the resulting PPI network was visualized using Cytoscape. The CytoHubba plugin identified key proteins based on their degree of interaction. Additionally, core subnetworks were extracted using the MCODE plugin with specific parameters. 2.5. Prediction of target genes of DEGs We utilized TargetScan (TargetScan:[79]http://targetscan.org/), miRanda (miRanda:[80]http://microrna.org/microrna/), and Starbase (Starbase, [81]https://starbase.sysu.edu.cn/) to predict target genes, focusing on the interactions of miRNA, mRNA, and lncRNA. Predictions were refined by intersecting results from these platforms, ensuring accuracy. A Venn diagram was generated to visualize common target genes, which were subsequently included in further analyses. Target gene prediction websites. 2.6. Construction of ceRNA regulatory network We utilized Targetscan and miRanda to predict target binding relationships between miRNA, mRNA, and lncRNA. Specifically, we selected DEGs with Targetscan scores ≥50, and miRanda energy 0.1. According to the competitive endogenous RNA (ceRNA) hypothesis, DElncRNAs and DEmRNAs within DEGs are expected to exhibit similar expression trends, while DEmiRNAs should show opposite trends [[82]26,[83]27]. Using these principles, we constructed differential expression lncRNA-miRNA-mRNA ceRNA regulatory networks and visualized them using Cytoscape software. 2.7. Survival and expression analysis of DEGs We performed survival analysis using the Kaplan-Meier Plotter database ([84]http://kmplot.com), which evaluates gene expression across 21 cancer types [[85]28]. This database assessed the prognosis of differentially expressed mRNAs, miRNAs, and lncRNAs identified in our study. Additionally, we utilized GEPIA (Gene Expression Profiling Interactive Analysis), integrating data from TCGA to analyze DEG expression across tissues [[86]29]. UALCAN is a TCGA-based resource for further expression analysis and biomarker identification [[87]30]. Proteinatlas provided insights into gene and protein expression in various tissues. These databases collectively informed us about DEG expression disparities between breast cancer and normal tissues. 2.8. Cell culture and transfection The MCF-7 (SCSP-531) and MDA-MB-231 (SCSP-5043) cell lines, representing two prevalent and highly proliferative breast cancer subtypes, along with the MCF-10A (SCSP-575) cell line, which serves as a model for normal breast epithelial cells, were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in high-glucose Dulbecco's Modified Eagle's Medium supplemented with 10 % fetal bovine serum (JYK-FBS-301, Jin Yuan Kang Biotechnology) at 37 °C with 5 % CO[2]. Transient transfections of specific gene-targeting siRNA fragments were achieved using Lipofectamine 3000 (Invitrogen). The designed siRNA sequences were validated for their knockdown efficiency, particularly targeting SNHG16, to elucidate its role in breast cancer cells. Gene sequences are listed in [88]Table S2. 2.9. Validation of DEG expression by real-time quantitative PCR Total RNA was extracted from MCF-7 and MDA-MB-231 cell samples using the TRIzol method, and RNA concentration and purity were assessed using NanoDrop 2000. RNA with concentrations ranging from 200 to 2000 ng/μl and an OD260/OD230 ratio of 1.8–2.2 was considered suitable. RNA was reverse transcribed into cDNA according to kit instructions. SYBR Green Mix (QP031, GeneCopoeia), primers, and gene templates were combined for amplification, with GAPDH as the reference gene for normalization. Data were analyzed using the 2^-ΔΔCT method with GraphPad Prism 8.0 software to quantify relative gene expression levels. Gene sequences are listed in [89]Table S3. 2.10. Western blotting experiment Cells were rinsed with PBS to remove the culture medium. RIPA lysis buffer (20 μl) was added per culture dish to achieve complete cell digestion. Additionally, 2 μl of protease inhibitor and 2 μl of phosphatase inhibitor (Thermo Scientific) were included. Protein concentration was measured using the BCA method. Samples were prepared with 1x loading buffer, boiled, and stored at −20 °C. SDS-PAGE gel electrophoresis was performed to separate proteins by size. Proteins were then transferred to PVDF membranes. A 5 % BSA solution incubation blocked nonspecific binding sites. Specific monoclonal antibodies were used for β-actin (1:1500) and AURKA (1:1000, EM1706-71, HUABIO), followed by overnight incubation at 4 °C. Secondary antibodies (Beyotime) conjugated with horseradish peroxidase-linked rabbit anti-mouse IgG (1:5000) were used, followed by exposure and imaging using the Epizyme Omni-ECL chromogenic reagent. ImageJ software was employed for film scanning to quantify density and determine the relative expression of target genes compared to β-actin. 2.11. Scratch assay Cells were enzymatically digested and seeded in 6-well plates until reaching 80 %–90 % confluency. Using a sterile, enzyme-free 10 μl pipette tip, scratches were made on the cell culture surface, ensuring uniform width and placement under a microscope. The old medium was replaced with serum-free DMEM and incubated for 24 h before observation and photography under low magnification. Cell migration rates were analyzed using ImageJ software. 2.12. CCK-8 assay Absorbance readings measured cell viability at 24, 48, and 72 h. Cells were digested with trypsin, washed with PBS, and seeded uniformly in 96-well plates (200 μl medium and 10 μl CCK-8 solution per well, NEST Biotechnology). After 4 h of incubation at 37 °C, absorbance at 450 nm was measured using a microplate reader to calculate cell proliferation rates. 2.13. Statistical analysis Sequencing data were analyzed and visualized using R 4.13. All other data were analyzed using SPSS 25.0, and graphs were drawn using GraphPad Prism 8.0.2. Quantitative data are presented as mean ± standard deviation. Statistical analysis employed ANOVA with a significance level set at α = 0.05 (two-tailed). 3. Results 3.1. Differential gene expression analysis We selected six breast cancer whole-genome expression datasets from the GEO database (lncRNA: [90]GSE65194, [91]GSE61304; miRNA: [92]GSE41922, [93]GSE45666; mRNA: [94]GSE29431, [95]GSE42568) based on predetermined criteria. To enhance the reliability of our differential gene selection, we intersected these results with those from the TCGA database. As illustrated in [96]Fig. 1A–I, upregulated genes (log[2] fold change (FC)>|2|, P < 0.05) are shown in red, downregulated genes (log[2] fold change (FC)>|2|, P < 0.05) in green, and non-significant genes in black. Upon intersection, we identified 112 DElncRNAs in tumor tissues compared to normal tissues, comprising 48 upregulated and 64 downregulated ones; 11 DEmiRNAs with 3 upregulated and 8 downregulated; and 97 DEmRNAs with 47 upregulated and 50 downregulated ([97]Fig. 1J–L). Fig. 1. [98]Fig. 1 [99]Open in a new tab Screening for differentially expressed genes. A, D, G: Differentially expressed lncRNA; B, E, H: Differentially expressed miRNA; C, F, I: Differentially expressed mRNA. J: Intersection of differentially expressed lncRNA; K: Intersection of differentially expressed miRNA; L: Intersection of differentially expressed mRNA. 3.2. Functional enrichment analysis of differential gene expression Functional enrichment analysis of DElncRNA-targeted genes revealed significant enrichment in protein binding, cell division, and cytoplasmic processes in GO analysis. KEGG pathway analysis highlighted enrichment in ECM receptor interaction, oocyte meiosis, Wnt/β-catenin signaling pathway, and cell cycle. For DEmiRNAs, GO analysis indicated enrichment in extracellular vesicle and membrane components, while KEGG pathways enriched in tyrosine metabolism, PPAR, and AMPK signaling pathways were noted. DEmRNA analysis showed GO enrichment in negative regulation of gene expression, positive regulation of cell cycle, and negative regulation of hematopoietic stem cells, with KEGG pathways enriched in Wnt/β-catenin signaling and immune system development ([100]Fig. 2). Fig. 2. [101]Fig. 2 [102]Open in a new tab Functional enrichment analysis of GO and KEGG for differential genes and their target genes. A: GO enrichment analysis of different expressed lncRNA target genes. B: KEGG enrichment analysis of different expressed lncRNA target genes. C: GO enrichment analysis of different expressed miRNA target genes. D: KEGG enrichment analysis of different expressed miRNA target genes. E: GO enrichment analysis of differently expressed mRNA. F: KEGG enrichment analysis of differently expressed mRNA. 3.3. Protein-protein interaction (PPI) network analysis Using the STRING database, we constructed PPI networks for DEmRNAs, filtering out isolated nodes. Separate networks were built for upregulated (47 nodes, 987 edges) and downregulated genes (50 nodes, 132 edges). Further analysis using Cytoscape software plugins MCODE and cytoHubba identified top-scoring core proteins in each network. Key genes included AURKA, HMMR, FN1, RUNX2, CDK1, CCNB1, CENPA, UHRF1, COL1A1, NUSAP1 for upregulated genes, and FABP4, LEP, SFRP1, FMO2, OGN, ADH1B, GPIHBP1, ADAMTS5, TGFBR3, ZBTB16 for downregulated genes. Notably, AURKA was central in the PPI network, and the top 20 genes by score were selected for further analysis ([103]Fig. 3A–C). Fig. 3. [104]Fig. 3 [105]Open in a new tab PPI network and key sub-network of differentially expressed mRNAs. A: PPI network of up-regulated differential genes. B: PPI network of down-regulated differential genes. C: Key sub-networks. D: CeRNA regulatory network. Red represents up-regulated differentially expressed genes, green represents down-regulated differentially expressed genes. The rectangle represents mRNA, the circle represents miRNA, and the prism represents lncRNA. E: Complementary sequences between SNHG16 and hsa-let-7b-5p, AURKA and hsa-let-7b-5p. 3.4. Interaction network and ceRNA network construction Using TargetScan and miRanda software, we predicted target genes for DElncRNAs and DEmiRNAs, intersecting these results with differentially expressed genes. This approach yielded 13 DElncRNAs, 3 DEmiRNAs, and 3 DEmRNAs. According to the ceRNA hypothesis, these lncRNAs should exhibit a negative correlation with miRNAs and a positive correlation with mRNAs. Constructing a breast cancer ceRNA regulatory network, we identified 29 interaction pairs, including 13 lncRNA-miRNA pairs, 13 lncRNA-mRNA pairs, and 3 miRNA-mRNA pairs ([106]Fig. 3D). Each adjacent differential gene pair exhibited opposite expression and prognostic relationships. Using the StarBase database, we further evaluated the correlation (R) strength between differential genes, focusing on the most robust ceRNA subnetwork (SNHG16-hsa-let-7b-5p-AURKA) for subsequent experimental validation ([107]Table S4). 3.5. Differential gene interactions in ceRNA regulatory networks Using RNA chip technology, we identified complementary binding sites between lncRNA SNHG16 and miRNA hsa-let-7b-5p ([108]Fig. 3E). This suggests SNHG16's potential involvement in the ceRNA regulation of hsa-let-7b-5p in breast cancer. Additionally, hsa-let-7b-5p was found to interact with mRNA AURKA, indicating its role in regulating mRNA expression via ceRNA mechanisms. 3.6. Expression of differential genes in ceRNA networks in breast cancer To validate ceRNA differential gene expression in breast cancer, we evaluated the prognostic value of SNHG16 and AURKA and hsa-let-7b-5p using the Kaplan-Meier database, and we assessed their expression using the UALCAN database ([109]Fig. 4A–C). To compare the expression of AURKA, we selected breast tissue sections from normal individuals (CAB001454, ID: 278) and breast cancer patients (HPA002636, ID: 2174) registered in the Proteinatlas database ([110]Fig. 4D). All results showed elevated SNHG16 and AURKA expression, and decreased hsa-let-7b-5p expression in breast cancer, correlating with prognosis and supporting ceRNA network mechanisms. Fig. 4. [111]Fig. 4 [112]Open in a new tab Differential gene expression. A: Survival analysis of AURKA in breast cancer prognosis. B: Survival analysis of SNHG16 in breast cancer prognosis. C: Survival analysis of hsa-let-7b-5p in breast cancer prognosis. D: AURKA expression in normal and breast cancer tissue sections. E: Expression of differential genes in breast cancer cells and normal breast epithelial cells (n = 4). ∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001. 3.7. qRT-PCR experimental results 3.7.1. qRT-PCR validation of differential gene expression in breast cancer cells We performed qRT-PCR to detect SNHG16, hsa-let-7b-5p, and AURKA expression in two breast cancer cell lines (MCF-7 and MDA-MB-231) and normal breast epithelial cells (MCF-10A). As shown in [113]Fig. 4E, SNHG16 and AURKA were significantly upregulated in breast cancer cells, while hsa-let-7b-5p was downregulated. SNHG16 showed more pronounced upregulation in MDA-MB-231 cells, and hsa-let-7b-5p downregulation was more evident in these cells, consistent with our predictions. 3.7.2. Knockdown of SNHG16 and qRT-PCR validation of gene knockdown efficiency To further investigate the role of lncRNA in ceRNA regulation, we designed si-SNHG16 interference fragments to knock down SNHG16 expression in cells. Fluorescence microscopy confirmed transfection efficiency above 90 %, indicating successful transfection for subsequent experiments. qRT-PCR revealed a significant reduction in SNHG16 expression post-transfection in MCF-7 cells, accompanied by decreased AURKA expression and increased hsa-let-7b-5p expression. Similar trends were observed in MDA-MB-231 cells, with notably increased hsa-let-7b-5p expression ([114]Fig. 5A–C). Fig. 5. [115]Fig. 5 [116]Open in a new tab Effect of knockdown SNHG16 expression on ceRNA. A: Efficiency of transfection of siRNA under fluorescence microscopy. B: qRT-PCR detection of transfection efficiency of si-SNHG16 on MCF-7 cells (n = 3). C: qRT-PCR detection of transfection efficiency of si-SNHG16 on MDA-MB-231 cells (n = 3). D, E: Effect of knockdown of SNHG16 on proliferation ability of MCF-7 and MDA-MB-231 in breast cancer cells (n = 4). ∗P < 0.05, ∗∗P < 0.01. 3.8. Impact of SNHG16 knockdown on proliferation and migration of breast cancer cells 3.8.1. Inhibition of breast cancer cell proliferation by SNHG16 knockdown CCK-8 assays demonstrated that knocking down SNHG16 inhibited breast cancer cell proliferation ([117]Fig. 5D–E). The most significant inhibition was observed at 48 h, indicating SNHG16's role in promoting breast cancer proliferation in both MCF-7 and MDA-MB-231 cells. 3.8.2. Inhibition of breast cancer cell migration by SNHG16 knockdown Scratch assays showed that SNHG16 knockdown suppressed breast cancer cell migration ([118]Fig. 6A–B). This further supports SNHG16's role in promoting breast cancer cell migration. Fig. 6. [119]Fig. 6 [120]Open in a new tab Effect of lncRNA SNHG16 on cell migration ability. A: Effect of knockdown of SNHG16 expression on breast cancer cell migration. B: Inhibition rate of breast cancer cell migration by knockdown of SNHG16 expression (n = 3). C. AURKA protein expression after knockdown of SNHG16 in both cell lines (n = 3). ∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001, ns represents p-value is not significant. 3.9. Verification of AURKA protein expression by western blotting To validate the expression of AURKA protein following the knockdown of lncRNA SNHG16 in MCF-7 and MDA-MB-231 cells, Western blot experiments were conducted. The results indicated a significant decrease in AURKA protein expression upon SNHG16 knockdown in both cell lines ([121]Fig. 6C). These findings corroborate with results from the GEPIA database and qRT-PCR analysis, confirming SNHG16's role in modulating AURKA protein expression through ceRNA mechanisms. This regulatory pathway is pivotal in influencing breast cancer cell proliferation and migration, thereby impacting disease progression and prognosis. 4. Discussion Breast cancer is the most prevalent tumor among women globally and ranks as the second most common cancer worldwide. Incidence and mortality rates are increasing annually [[122]1]. The disease often begins subtly, making early detection challenging, with 90 % of breast cancer patients succumbing to metastasis [[123]31]. In this study, we conducted gene screening using TCGA and GEO databases to identify differentially expressed genes in breast cancer. Functional enrichment analyses of these genes and their targets revealed significant involvement in cell division-related activities, underscoring the high metastatic potential of breast cancer. SNHG16 was initially detected to be highly expressed in invasive neuroblastomas [[124]32]. Recent experiments and sequencing results have consistently shown elevated SNHG16 expression in various cancers such as liver, colorectal, and breast cancers, aligning with our expectations [[125][33], [126][34], [127][35]]. Studies indicate that high SNHG16 expression contributes to breast cancer progression through multiple mechanisms, including promoting cell proliferation, metastasis, angiogenesis, and chemoresistance [[128][36], [129][37], [130][38], [131][39], [132][40]]. In breast cancer, SNHG16 was newly found to act as a novel biomarker through ceRNA interactions [[133][41], [134][42], [135][43], [136][44]]. AURKA positively regulates the G2 to M transition in the cell cycle and spindle-related activities during early mitosis [[137]44]. Its interactions with substrates like histone H3 and proteins involved in centrosome separation and spindle formation underscore its essential role in mitotic progression, highlighting its role in breast cancer progression [[138][45], [139][46], [140][47]]. For example, AURKA collaborates with HMMR in spindle assembly during mitosis [[141][48], [142][49], [143][50]], while CDK1 stabilizes AURKA expression, enhancing its role in cell cycle regulation [[144]51]. The reliability of these core differential genes and subnetworks was validated through multiple experimental studies, emphasizing their critical roles in cancer progression. However, comprehensive studies integrating large-scale gene datasets and experimental validation are needed to elucidate their roles in breast cancer progression. Integration of predicted target genes yielded a final set of differentially expressed genes, forming a ceRNA regulatory network. We identified SNHG16-hsa-let-7b-5p-AURKA as the most robustly correlated ceRNA network in breast cancer. Previous studies have validated SNHG16's role as an upstream regulator of hsa-let-7b-5p in hepatocellular carcinoma [[145]52]. In contrast, AURKA's downstream influence on hsa-let-7b-5p has been observed in neuroglial tumors [[146]53], affirming the regulatory presence of this ceRNA in cancer control. However, the potential role of this regulatory axis in breast cancer has not been demonstrated. Prognostic analysis of DEGs within the ceRNA network in breast cancer revealed high expression levels of SNHG16 and AURKA correlating with poor prognosis, whereas low hsa-let-7b-5p expression indicated unfavorable outcomes. There have been experimental validations have confirmed AURKA's overexpression in ER-positive and triple-negative breast cancers, which is similar to our experimental results [[147]54,[148]55]. In contrast, low hsa-let-7b-5p levels correlate with poorer prognosis in breast cancer, possibly due to its negative correlation with SMUG1 expression, impacting survival rates in estrogen receptor-positive breast cancer [[149]56]. SNHG16 is implicated in several pathways to modulate tumorigenesis; for instance, downregulating SNHG16 in esophageal cancer inhibits the Wnt/β-catenin pathway, thereby suppressing cell proliferation and invasion [[150]57]. Similarly, reduced SNHG16 expression in bladder cancer regulates downstream proteins (Wnt1, c-myc, cyclin-D1, and catenin) through the Wnt/β-catenin pathway to inhibit T24 cell proliferation and migration [[151]58]. SNHG16 also enhances TGF-β1/SMAD5 pathway activity via the miR-16-5P and SMAD5 ceRNA axis, promoting breast cancer development [[152]59]. We experimentally confirmed the impact of SNHG16 downregulation on cell proliferation and migration. Our findings revealed varying inhibition in MCF-7 and MDA-MB-231 cell lines, consistent with previous studies [[153]59,[154]60]. Specifically, MDA-MB-231 cells showed pronounced inhibition of proliferation, while MCF-7 cells exhibited greater suppression of migration, likely due to differences in cellular morphology and growth rates, necessitating further experimental validation. However, our study did not analyze the expression of differential genes across all breast cancer subtypes or the effect on breast cell proliferation after the knockout of other genes on this axis, warranting further exploration. 5. Conclusion In conclusion, we successfully constructed a prognostically relevant ceRNA regulatory network in breast cancer through differential gene screening. Experimental validation confirmed SNHG16-hsa-let-7b-5p-AURKA in breast cancer cells and affirmed that SNHG16 promotes cell proliferation and migration. CRediT authorship contribution statement Zipeng Qiao: Writing – original draft, Visualization, Validation, Software, Methodology, Conceptualization. Yongjun Tang: Validation, Software, Methodology, Data curation, Conceptualization. Shengle Li: Visualization, Validation, Software. Qiufeng Lao: Software, Methodology, Investigation, Conceptualization. Yu Xing: Visualization, Software, Conceptualization. Qingquan Zhang: Methodology, Investigation, Formal analysis. Songying Pan: Validation, Methodology. Chaoyong Bei: Visualization, Resources, Investigation. Weiyi Pang: Writing – review & editing, Project administration, Funding acquisition, Conceptualization. Hui Liu: Writing – review & editing, Supervision, Project administration, Conceptualization. Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Acknowledgments