Abstract Background Hyperoside (Hyp) exerts considerable inhibitory effects on non-small-cell lung cancer (NSCLC) cells. However, the underlying molecular mechanisms have not been fully elucidated. We aimed to explore the molecular mechanisms of Hyp in the treatment of NSCLC using a combination of network pharmacology and in vitro experiments. Methods Active targets of Hyp and NSCLC-related targets were identified using public databases. The therapeutic targets were predicted based on the intersection of drug and disease targets. A protein–protein interaction (PPI) network was constructed using STRING and Cytoscape, and key network modules and targets were identified through network topology analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using DAVID. Molecular docking between Hyp and therapeutic targets was performed using AutoDock Vina software. These results were further verified by in vitro experiments of A549 cell lines, including the CCK-8 assay, EdU staining, flow cytometry and western blotting. Results A total of 30 therapeutic targets of Hyp against NSCLC were identified. Analysis of the PPI network revealed a key network module and five key targets: MMP9, CASP3, MAPK1, ESR1, and EGFR. These targets were related to cell proliferation and migration, apoptosis, and oxidative stress and may play a role through the FoxO, MAPK, Rap1, Ras, and PI3K-Akt signaling pathways. Molecular docking results showed a strong binding affinity between Hyp and most therapeutic targets. Further experiments confirmed that Hyp inhibited proliferation and induced apoptosis of A549 cells by regulating the EGFR/ERK/FOXO1 signaling pathway. Conclusions Hyp may be a promising drug for treating NSCLC (especially lung adenocarcinoma), and its therapeutic mechanisms is closely related to the regulation of EGFR/ERK/FOXO1 pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-03622-9. Keywords: Hyperoside, Non-small-cell lung cancer, Network pharmacology, Apoptosis, EGFR/ERK/FOXO1 pathway Introduction According to the latest statistics from the International Agency for Research on Cancer (IARC) in 2022, the incidence and mortality rates of lung cancer in the world have risen to the highest among malignant tumors [[36]1]. Based on histopathology, lung cancer can be divided into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLC accounts for 85% of all lung cancer cases [[37]2]. NSCLC cells are characterized by the slow growth and division, resulting in late diffusion and metastasis. Thus, their early stage is not easy to detect [[38]3]. In recent years, great progress has been made in the surgical or non-surgical treatment of NSCLC. However, its clinical treatment and prognosis are still poor, and the 5-year survival rate of NSCLC is approximately 15% [[39]4, [40]5]. Clinical studies have shown that a great number of traditional Chinese medicine treatments have exhibited satisfactory anti-tumor effects. Meanwhile, compared to traditional chemotherapy drugs, traditional Chinese medicines possess fewer adverse reactions and lower drug resistance [[41]6, [42]7]. Hyperoside (Hyp) is a natural flavonol glycoside that is widely found in a variety of plants such as Hypericum monogynum, Crataegus pinnatifida, and Polygonum aviculare [[43]8]. It has been found to own various biological activities such as anti-inflammatory, antibacterial, antiviral, and organ protection [[44]9]. Currently, Hyp has also been found to exert considerable inhibitory effects on NSCLC cells. It was reported that Hyp could inhibit the proliferation and induce the apoptosis of T790M-positive NSCLC cells. The further studies revealed that this effect is closely associated with the upregulated expression of forkhead box O1 (FOXO1) [[45]10]. It was also demonstrated that Hyp could activate the p38/MAPK and JNKinduced mitochondrial death pathway, remarkably accelerating the A549 cells apoptosis [[46]11]. Besides these, another study disclosed that Hyp could trigger the apoptosis of A549 cell by enhancing autophagy [[47]12]. In brief, Hyp may exerts anti-NSCLC effects through a variety of biological mechanisms. Hence, It is important and necessary to further clarify its molecular mechanisms. Traditional methods for screening the pharmacological activities of natural monomer components are often achieved by cell experiments or Qualcomm’s method. However, these research methods are time-consuming and expensive, and their accuracy is closely related to the overall conditions of animal models or human bodies [[48]11]. Network pharmacology is an emerging discipline that integrates systems biology, molecular biology, pharmacology, and various computing platforms in the era of big data. It could perform multi-level network screening and construction from the macro to micro perspective, visualize the data using a variety of database platforms and computer software, and more intuitively explain the complex relationship between traditional Chinese medicine and diseases. Network pharmacology emphasizes the understanding of the interaction between drugs and the organism from the perspective of biological network balance, and explains the occurrence and development process of disorders, which is consistent with the characteristics of holistic treatment of traditional Chinese medicine. Its rapid development has also brought new ideas to the field of traditional Chinese medicine [[49]12, [50]13]. This study used network pharmacology combined with in vitro biological experiments to explore the molecular mechanisms of Hyp in the treatment of NSCLC. A flowchart of this study is summarized in Fig. [51]1. Fig. 1. [52]Fig. 1 [53]Open in a new tab Detailed flowchart of this study Materials and methods Network pharmacological analysis Prediction of Hyp active targets The SMILE file and 3D structure file in SDF format of Hyp were downloaded using “hyperoside” as the keyword in PubChem [[54]14] ([55]https://pubchem.ncbi.nlm.nih.gov/). In Traditional Chinese Medicine Systems Pharmacology (TCMSP) [[56]15] ([57]https://old.tcmsp-e.com/tcmsp.php), the active targets of Hyp were obtained using the “hyperoside” term. Potential targets were also predicted using the SMILE file in Search Tool for Interacting Chemicals (STITCH) [[58]16] ([59]http://stitch.embl.de/). In addition, the SDF format file was uploaded to PharmMapper [[60]17] ([61]http://www.lilab-ecust.cn/pharmmapper/) for reverse molecular docking simulation to screen out the active targets of Hyp. In PharmMapper, the species was set as “homo sapiens”. The predicted targets with fit score ≥ 2.5 from PharmMapper were collected in our study. Acquisition of NSCLC disease targets NSCLC disease targets were collected using the keywords “non-small-cell lung cancer ” in Online Mendelian Inheritance in Man (OMIM) [[62]18] ([63]https://omim.org/), Therapeutic Target Database (TTD) [[64]19] ([65]http://db.idrblab.net/ttd/), Comparative Toxicogenomics Database (CTD) [[66]20] ([67]http://ctdbase.org/), and DisGeNET [[68]21] ([69]https://www.disgenet.org/). The above NSCLC disease targets were standardized using UniProt [[70]22] ([71]https://www.uniprot.org/) and duplicate proteins were removed. The proteins with “direct evidence” as “marker or mechanism or therapeutic” in CTD were selected as the NSCLC disease targets that had more sufficient evidence. Prediction of therapeutic targets of Hyp against NSCLC The active targets of Hyp and NSCLC disease targets were intersected using Venny (version 2.1.0) ([72]https://bioinfogp.cnb.csic.es/tools/venny/index.html) to obtain the potential therapeutic targets of Hyp against NSCLC. Construction and analysis of PPI network STRING [[73]23] ([74]https://string-db.org/) was used to get the interactions between therapeutic targets by setting the species as “homo sapiens” and the confidence score ≥ 0.4. These interactions were then imported into Cytoscape [[75]24] (version 3.2.0) to construct a PPI network. The MCODE plugin [[76]25] with default parameters from Cytoscape was used to identify the key network modules. The MCODE could detect the densely connected regions in the PPI network, and the network module with the highest score was regarded as the key network module. The degree value of the nodes from the key network module was analyzed with the plugin Network Analyzer [[77]26], and the key targets were then identified according to the screening criteria that the degree value was greater than or equal to its average value. The degree value of a node reflects the number of connections with the other nodes in the network. The greater its value, the greater its importance to the entire network. Therefore, the degree in this study was selected as an important index for identifying key targets. GO and KEGG pathway enrichment analyses Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses for the target proteins in the key network module were performed using Database for Annotation, Visualization, and Integrated Discovery (DAVID) [[78]27] ([79]https://david.ncifcrf.gov/). The screening condition (p < 0.01) was used to obtain significantly enriched GO items and KEGG pathways, and the results were visualized using the R software. Construction of “Hyp-Target-Pathway” network Firstly, the above obtained Hyp-target interaction relationships were constructed into a “Hyp-Target” network by Cytoscape, and then the corresponding relationships of target-pathway were built into a “Target-Pathway” network by the same way. Finally, the two networks were integrated into a “Hyp-Target-Pathway” network using the merge function of Cytoscape. Molecular docking simulation The crystal structure files of the targets in the key network module were accessed from the Research Collaboratory for Structural Bioinformatics Protein Data Bank (RCSB PDB) [[80]28] ([81]http://www.rcsb.org/), and the PyMOL software [[82]29] was employed to remove water molecules and original ligands from the protein structures. The SDF format of Hyp was converted into the PDB format using OpenBabel software [[83]30]. Using AutoDock Tools (version 1.5.6), the structural files of Hyp and the targets were hydrogenated, charged, and saved in the PDBQT format. Molecular docking was performed using AutoDock Vina [[84]31] on a Linux platform, and the docking results were visualized using PyMOL. In vitro experimental verification Cell culture The NSCLC cell line A549 was procured from the Shanghai Cell Bank of the China Academy of Sciences. A549 cells were cultured in RPMI 1640 medium (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 100 µg/ml streptomycin, 10% fetal bovine serum, and 100 U/ml penicillin. The cells were routinely subcultured in an incubator at 37 °C and 5% CO[2]. Cytotoxicity test Hyp (98.68% purity) was purchased from MedMol (Shanghai, China). Before use, Hyp was dissolved in an appropriate volume of dimethyl sulfoxide and diluted to the required concentrations. A549 cells in the logarithmic growth phase were seeded in 96-well plates at a density of 5 × 10^3 cells/well and cultured for 24 h. The A549 cells in the drug group were treated with different concentrations of Hyp solution (20, 40, 80, 160, and 320 µM) for 24, 48, and 72 h, whereas cells in the control group were treated with drug-free medium for 24, 48, and 72 h. Subsequently, 10 µL of CCK-8 reagent (Beyotime, Shanghai, China) was added to each well and incubated for another 2 h. The absorbance was measured at 490 nm using a microplate reader, and the cell survival rate was calculated. This experiment was conducted to screen for the optimal drug concentration that interfered with cell viability. Cell proliferation test A549 cells were seeded into 6-well plates at a density of 2 × 10^5 cells/mL per well and cultured for 24 h. In the Hyp group, 160 µM Hyp solution was added; in the Hyp + NSC228155 (specific EGFR activator) (MedMol, Shanghai, China) group, 160 µM Hyp solution and 2 µM NSC228155 were added; and in the control group, a drug-free medium was used. The cultures were maintained for an additional 24 h. Then, each group was treated with 10 µM EdU (Ribobio, Guangzhou, China) at 37℃ for 2 h. Subsequently, A549 cells in each group were incubated with Click reaction solution (Meilunbio, Dalian, China) for 30 min at room temperature, washed three times with phosphate buffered saline (PBS), and stained with 10 µg/mL DAPI (Meilunbio, Dalian, China) for 10 min. Finally, the cells were observed and photographed under an inverted fluorescence microscope. The resulting images were processed using ImageJ software. Apoptosis detection A549 cells were seeded into 6-well plates and treated with Hyp or NSC228155 for 24 h. The grouping and administration of cells followed the procedure described in Sect. [85]2.2.3. The cells were then digested with trypsin, washed twice with PBS, centrifuged at 1500 rpm for 5 min, and resuspended in 100 µL binding buffer. Thereafter, the cells were stained with 5 µL annexin V-FITC (Beyotime, Shanghai, China) and 5 µL PI (Beyotime, Shanghai, China) for 15 min at room temperature. Apoptosis was detected by flow cytometry. Western blotting analysis The grouping and administration of cells followed the same procedure as described in Sect. [86]2.2.3. After 24 h of treatment with Hyp or NSC228155, RIPA lysis buffer (Beyotime, Shanghai, China) was added to each group of cells to prepare total protein lysates. Then the BCA protein assay kit (Beyotime, Shanghai, China) was used for protein quantification. Protein samples were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, transferred to nitrocellulose membranes, blocked with 5% skimmed milk (Beyotime, Shanghai, China) for 2 h at room temperature, and incubated with primary antibodies against epidermal growth factor receptor (EGFR), p-EGFR, extracellular regulated kinase 1/2 (ERK1/2), p-ERK1/2, FOXO1, Cleaved-Caspase-3, Bax, Bcl-2, or β-actin overnight at 4℃. The primary antibodies EGFR, p-EGFR, ERK1/2, p-ERK1/2, FOXO1, and β-actin were purchased from Abcam (Cambridge, MA, USA) and Cleaved-Caspase-3, Bax, and Bcl-2 were purchased from Cell Signaling Technology (Danvers, MA, USA). The membrane was washed with TBST and incubated with a secondary antibody at room temperature for 2 h. The protein bands were detected using enhanced chemiluminescence detection reagents (Danvers, MA, USA). β-Actin was used as a control to standardize the expression of other proteins. Band intensity was analyzed using ImageJ software. Statistical analysis All results are presented as the mean ± standard deviation (SD), and the data were analyzed using GraphPad Prism software (version 8.0). Differences between two groups were measured using t-test. One-way analysis of variance (ANOVA) was used to assess the statistical differences between multiple groups. For pairwise comparisons, Fisher’s least significant difference (LSD) test was applied. Differences were considered statistically significant at p < 0.05. Results Therapeutic targets of Hyp against NSCLC Seven, five, and 265 active Hyp targets were obtained from the TCMSP, STITCH, and PharmMapper databases, respectively. After removing duplicate targets, 273 unique active targets were identified. Three, 82, 143, and 151 NSCLC disease targets were retrieved from the OMIM, TTD, CTD, and DisGeNET, respectively. In total, 344 NSCLC disease targets were collected after removing duplicates. Venny was utilized to intersect the Hyp active targets with the NSCLC disease targets and draw a Venn diagram (Fig. [87]2A). Eventually, 30 potential therapeutic targets of Hyp acting on NSCLC were identified. Fig. 2. [88]Fig. 2 [89]Open in a new tab Network analysis of the potential therapeutic targets against NSCLC. A The Venn diagram of Hyp targets and NSCLC targets. B The network module analysis of PPI network. C The topology analysis of network module 1 (node size representing degree value) PPI network, key network modules, and key targets The PPI network for Hyp treating NSCLC was successfully generated through Cytoscape (Fig. [90]2B). This network contained 30 nodes and 128 edges, in which nodes represented proteins, and edges represented the interaction between proteins. Two network modules were identified using the MCODE plugin from Cytoscape (Fig. [91]2B). The score of network module 1 was 9.273, including 12 nodes and 51 edges. The score of network module 2 was 4.000, including four nodes and six edges. The score of network module 1 was much larger than that of network module 2. Thus, network module 1 was regarded as the key network module of the PPI network. Network Analyzer was employed to calculate the degree value of the nodes in network module 1, and the results were shown in Table [92]1. The degree values of matrix metalloproteinase-9 (MMP9), caspase-3 (CASP3), mitogen-activated protein kinase 1 (MAPK1), estrogen receptor (ESR1) and EGFR were 11, 11, 11, 11 and 9, respectively, which were all greater than the average value (8.5). Thus, they were considered to be the key targets of network module 1. These proteins were also key targets of the whole PPI network and may play a crucial role in the biological effects of Hyp in NSCLC treatment. Table 1. The degree value of therapeutic targets in network module 1 Gene symbol Protein name Uniprot ID Degree value CASP3 Caspase-3 [93]P42574 11 ESR1 Estrogen receptor [94]P03372 11 MAPK1 Mitogen-activated protein kinase 1 [95]P28482 11 MMP9 Matrix metalloproteinase-9 [96]P14780 11 EGFR Epidermal growth factor receptor [97]P00533 9 IGF1R Insulin-like growth factor 1 receptor [98]P08069 8 CAT Catalase [99]P04040 7 KDR Vascular endothelial growth factor receptor 2 [100]P35968 7 MET Hepatocyte growth factor receptor [101]P08581 7 PGR Progesterone receptor [102]P06401 7 SOD2 Superoxide dismutase [Mn], mitochondrial [103]P04179 7 NQO1 NAD(P)H dehydrogenase [quinone] 1 [104]P15559 6 [105]Open in a new tab GO and KEGG pathway enrichment analyses A total of 50 significant items were screened by GO enrichment analysis, including 33 biological processes, four cellular components, and 13 molecular functions. The top 10 items of them were shown in Fig. [106]3A. Biological processes mainly included negative regulation of apoptosis process, positive regulation of cell proliferation or migration, response to hydrogen peroxide, etc. Cell components included membrane raft, golgi apparatus, plasma membrane, and focal adhesion. Molecular functions primarily included transmembrane receptor protein tyrosine kinase, protein tyrosine kinase activity, superoxide dismutase activity, etc. Fig. 3. [107]Fig. 3 [108]Open in a new tab Enrichment analysis of therapeutic targets and construction of “Hyp-Target-Pathway” network. A GO enrichment analysis. B KEGG pathway enrichment analysis. C “Hyp-Target-Pathway” network (Light blue triangle representing Hyp, green circle representing target, and square representing pathway) The results of KEGG pathway enrichment analysis exhibited that 32 biological pathways were significantly enriched (Fig. [109]3B). These pathways involved human diseases (such as proteoglycans in cancer, breast cancer, NSCLC, etc.), organismal system (such as estrogen signaling pathway, longevity regulating pathway, progesterone-mediated oocyte maturation, etc.), and signal transduction (such as FoxO signaling pathway, MAPK signaling pathway, Rap 1 signaling pathway, etc.). This study focused on the signal transduction pathway that was identified as responsible for environmental information processing in KEGG PATHWAY database. The p values of FoxO and MAPK signaling pathways were relatively small, which may be the key mechanisms of Hyp in treating NSCLC. The “Hyp-Target-Pathway” network was successfully established using Cytoscape (Fig. [110]3C). This network consisted of 36 nodes (one compound, 12 targets, and 23 pathways) and 100 edges. This indicated that Hyp owned the characteristics of multiple targets and pathways in the treatment of NSCLC. Molecular docking When the binding energy is less than − 7.0 kcal/mol, it indicates strong binding activity between the ligand and receptor [[111]32]. Except CASP3, ESR1, and PGR, the binding energies of other proteins with Hyp were all less than − 7.0 kcal/mol (Table [112]2), suggesting that the results obtained via network pharmacology were reliable.The binding energies of the key targets, namely, EGFR, MAPK1, MMP9, ESR1, and CASP3, were − 9.8, − 9.0, − 7.5, − 6.4, and − 5.1 kcal/mol, respectively. Hyp exhibited strong binding activity with EGFR, MAPK1, and MMP9. As shown in Fig. [113]4, these three complexes generated stable conformations through multiple hydrogen bonds. Table 2. Molecular Docking results between hyp and therapeutic targets in network module 1 Symbol Protein name PDB ID Binding energy(kcal/mol) CASP3 Caspase-3 3KJF − 5.1 CAT Catalase 1DGH − 9.3 EGFR Epidermal growth factor receptor 3UG2 − 9.8 ESR1 Estrogen receptor 4MGB − 6.4 IGF1R Insulin-like growth factor 1 receptor 5FXR − 7.9 KDR Vascular endothelial growth factor receptor 2 3U6J − 8.2 MAPK1 Mitogen-activated protein kinase 1 4FV5 − 9.0 MET Hepatocyte growth factor receptor 4MXC − 7.8 MMP9 Matrix metalloproteinase-9 2OW1 − 7.5 NQO1 NAD(P)H dehydrogenase [quinone] 1 5EA2 − 10.1 PGR Progesterone receptor 3KBA − 5.5 SOD2 Superoxide dismutase [Mn], mitochondrial 5T30 − 7.1 [114]Open in a new tab Fig. 4. [115]Fig. 4 [116]Open in a new tab The molecular docking between Hyp and key targets. MAPK1 mitogen-activated protein kinase 1, MMP9 matrix metalloproteinase-9, EGFR epidermal growth factor receptor Hyp inhibiting viability and proliferation of A549 cells After 24, 48, and 72 h of treatment with different concentrations of Hyp, the survival rate of A549 cells significantly decreased (p < 0.05). At the same intervention times, with an increase in Hyp concentration, the cell survival rate decreased (Fig. [117]5). At the same Hyp concentration, longer intervention times corresponded to lower cell survival rates (Fig. [118]5). These results showed that Hyp inhibited the activity of A549 cells in a time- and dose-dependent manner. The IC50 values for Hyp at 24, 48, and 72 h in this study were 149.41, 102.88, and 64.26 µM, respectively. Because the treatment with 160 µM Hyp for 24 h had a good inhibitory effect on A549 cells, this concentration was used for follow-up experiments. The EdU positivity rate in the Hyp group was significantly lower than that in the control group (p < 0.05; Fig. [119]6). Compared with the Hyp group, the EdU positivity rate in the Hyp + NSC228155 group was significantly higher (p < 0.05) (Fig. [120]6). Fig. 5. [121]Fig. 5 [122]Open in a new tab Hyp suppressed cell viability in A549 cells (n = 5). A549 cells were treated with different concentrations of Hyp (0-320µM) for 24 h, 48 h and 72 h, respectively. All data were expressed as mean ± SD. Differences were analyzed using t-test. ^****p < 0.0001 vs. control (0 µM group). ^#p < 0.05, ^##p < 0.01, ^###p < 0.001, ^####p < 0.0001 vs. 24 h Fig. 6. [123]Fig. 6 [124]Open in a new tab Hyp inhibited the proliferation of A549 cells (n = 3). EdU assay was used to detect the proliferation of A549 cells. All data were expressed as mean ± SD. Differences were analyzed using ANOVA. Fisher’s LSD test was used for pairwise comparisons. ^*p < 0.05, ^**p < 0.01 Hyp inducing apoptosis of A549 cells The effect of Hyp on the apoptosis of A549 cells was detected using flow cytometry. Our results (Fig. [125]7) showed that the rate of apoptosis in the Hyp group was significantly higher than that in the control group (p < 0.05). By contrast, the apoptosis rate in the Hyp + NSC228155 group was significantly lower (p < 0.05) than that in the Hyp group. Western blot results (Fig. [126]8) showed that compared with the control group, in the Hyp group, the expression of pro-apoptosis proteins Cleaved-Caspase-3 and Bax increased significantly (p < 0.05), whereas the expression of the anti-apoptotic protein Bcl-2 decreased significantly (p < 0.05). These results indicated that Hyp induced apoptosis in A549 cells. Fig. 7. [127]Fig. 7 [128]Open in a new tab Hyp induced the apoptosis of A549 cells (n = 3). The apoptosis of A549 cells was detected by flow cytometry. All data were expressed as mean ± SD. Differences were analyzed using ANOVA. Fisher’s LSD test was used for pairwise comparisons. ^**p < 0.01, ^***p < 0.001 Fig. 8. [129]Fig. 8 [130]Open in a new tab Hyp regulated apoptosis-related proteins in A549 cells (n = 3). The expression levels of apoptosis-related proteins were determined by Western blot. All data were expressed as mean ± SD. Differences were analyzed using ANOVA. Fisher’s LSD test was used for pairwise comparisons. ^*p < 0.05, ^**p < 0.01, ^***p < 0.001, ^****p < 0.0001 Hyp regulating EGFR/ERK/FOXO1 signaling pathway The expression levels of proteins related to the EGFR/ERK/FOXO1 signaling pathway were shown in Fig. [131]9. Compared to the control group, in the Hyp group, the p-EGFR/EGFR and p-ERK1/2/ERK1/2 ratios decreased significantly (p < 0.05), whereas the expression level of FOXO1 increased significantly (p < 0.05). By contrast, compared to the Hyp group, in the Hyp + NSC228155 group, the ratios of p-EGFR/EGFR and p-ERK1/2/ERK1/2 were significantly increased (p < 0.05), whereas the expression of FOXO1 was significantly downregulated (p < 0.05). These results suggested that Hyp inhibited the activities of EGFR and ERK1/2 and downregulated the expression of FOXO1. Fig. 9. [132]Fig. 9 [133]Open in a new tab Hyp regulated the EGFR/ERK/FOXO1 signaling pathway-related proteins in A549 cells (n = 3). The expression levels of related protein in the EGFR/ERK/FOXO1 signaling pathway were determined by western blot. All data were expressed as mean ± SD. Differences were analyzed using ANOVA. Fisher’s LSD test was used for pairwise comparisons. ^*p < 0.05, ^**p < 0.01, ^***p < 0.001. EGFR: epidermal growth factor receptor; ERK1/2: extracellular regulated kinase 1/2; FOXO1: forkhead box O1 Discussion NSCLC is a malignant tumor that seriously endangers human health, and its incidence is increasing annually. Despite the rapid development of medical technologies, the overall therapeutic effect on NSCLC remains unsatisfactory. Hyp is an important natural product that has attracted considerable attention in the medical field. Hyp, a flavonoid glycoside, complies with the five principles of Lipski, indicating that it has good pharmacokinetic properties [[134]8]. Hyp exerts strong inhibitory effects on NSCLC cells [[135]10, [136]33, [137]34]. However, the underlying therapeutic mechanisms have not been fully elucidated. Using a combination of network pharmacology and in vitro biological experiments, we explored the molecular mechanisms of Hyp treatment in NSCLC from the viewpoint of the drug–target-pathway. In this study, we identified 30 therapeutic targets using network pharmacology. After topological analysis of the PPI network, five key targets were identified: MMP9, CASP3, MAPK1, ESR1, and EGFR. These targets have a high degree value, indicating that they may play a crucial role in the mechanism of action of Hyp in the treatment of NSCLC. In addition, the results of the molecular docking analysis showed that Hyp had good binding activity with EGFR, MAPK1, and MMP9. The docking score between EGFR and Hyp was the lowest, indicating the highest binding affinity. EGFR is a cell surface receptor encoded by the ErbB1 gene, and its overexpression or abnormal activation is closely associated with the progression of NSCLC [[138]35]. Currently, receptor tyrosine kinase inhibitors such as gefitinib and erlotinib, which target EGFR, have been effectively used in the clinical treatment of NSCLC [[139]36]. MAPK1 is a downstream target of the MAPK signaling pathway, also known as ERK2, which can transfer extracellular information into cells, thus regulating the cell proliferation, apoptosis, and differentiation. Compared to normal cells, the noticeably downregulated expression of MAPK1 was detected in NSCLC cells [[140]37], and MAPK1 knockdown remarkably attenuated the proliferation and metastasis of lung adenocarcinoma cells [[141]38]. MMP9 is a vital enzyme in the human body that could specifically degrade the extracellular matrix or substrate and participates in the tumor cells invasion and metastasis. MMP9 was highly expressed that was positively correlated with the invasion progression of NSCLC cells [[142]39]. It was found that blocking the expression of MMP9 with a PI3K inhibitor could weaken their invasion and metastasis [[143]40]. As one of the most important executors of cell apoptosis, CASP3 is activated by upstream signals and can produce cascade effects to induce the apoptosis. One study confirmed by in vitro experiments that CASP3 was involved in the Hyp-induced apoptotic process in NSCLC cells [[144]34]. ESR1, an estrogen receptor, is closely associated with the occurrence, progression, and metastasis of breast cancer. In recent years, some studies have reported that ESR1 has clinical significance in the diagnosis, treatment, and prognosis of NSCLC [[145]41, [146]42]. Our GO enrichment analysis results showed that these potential targets were mainly involved in cell proliferation, migration, apoptosis, and oxidative stress, which were important factors influencing Hyp against NSCLC. KEGG pathway enrichment analysis showed that FoxO and MAPK signaling pathways were closely related to the molecular mechanisms of Hyp against NSCLC. MAPK1, EGFR, and insulin-like growth factor 1 receptor(IGF1R) were enriched in the FoxO and MAPK signaling pathways. Among these, MAPK1 and EGFR were key targets and showed satisfactory binding activity with Hyp. FOXO is a member of the forkhead transcription factor family, which includes four members: FOXO1, FOXO3, FOXO4, and FOXO6. They participate in many physiological and pathological processes such as apoptosis, the cell cycle, and DNA damage repair by regulating the expression of downstream genes [[147]43]. FOXO1 is underexpressed in NSCLC cells, and its upregulation is a favorable prognostic factor [[148]44, [149]45]. Hu et al. [[150]10] clarified that the anti-NSCLC activity of Hyp may be attributed to the inhibition of proliferation and apoptosis of tumor cells by upregulating FOXO1 expression. The AMPK signaling pathway has been confirmed to be located upstream of FOXO proteins, and its disruption could alter the activity or expression of FOXO protein [[151]46]. Based on these results, we suspect that Hyp regulates the proliferation and apoptosis of tumor cells by acting on the EGFR/ERK/FOXO1 pathway, thus playing a therapeutic role in NSCLC. In this study, the potential relationship between Hyp and NSCLC was systematically analyzed by network pharmacology method. To test this hypothesis, we conducted several in vitro experiments. Because A549 cell line was the most widely used adenocarcinoma model in the research of NSCLC, our study chose this cell line as the experimental object, aiming at clarifying the molecular mechanisms of Hyp in the treatment of NSCLC. The CCK-8 assay showed that Hyp reduced the viability of A549 cells in a time- and dose-dependent manner. We observed, through EdU staining, that Hyp effectively inhibited the proliferation of A549 cells and the EGFR activator NSC228155 attenuated this inhibition. We also found that after Hyp treatment, the apoptosis rate of A549 cells increased significantly, and the expression of Cleaved-Caspase-3 and Bax increased significantly, whereas the expression of Bcl-2 decreased significantly, indicating that Hyp could induce apoptosis in A549 cells. Notably, when Hyp and NSC228155 were used simultaneously, the apoptosis rate of A549 cells was significantly reduced. In addition, we studied the effect of Hyp on the EGFR/ERK/FOXO1 pathway using western blotting. The results showed that the ratios of p-EGFR/EGFR and p-ERK1/2/ERK1/2 decreased in A549 cells after Hyp intervention, suggesting that Hyp activates EGFR and ERK1/2 through protein phosphorylation. Following Hyp treatment, the expression level of FOXO1 in A549 cells increased significantly. However, NSC228155 counteracted these changes, indicating that Hyp upregulated FOXO1 expression by inhibiting the activities of EGFR and ERK1/2. Therefore, we believe that Hyp has the potential to treat NSCLC, primarily by regulating the EGFR/ERK/FOXO1 pathway to inhibit the proliferation of tumor cells and induce the apoptosis of tumor cells. In this study, several key targets and promising signaling pathways were successfully identified through network pharmacology. However, due to the limitation of research funds, the multi-target and multi-pathway mechanism of Hyp in NSCLC has not been systematically verified by experiments. Our in vitro experiment only used A549 adenocarcinoma cell line, but failed to cover other subtypes of NSCLC, such as squamous cell carcinoma, which may lead to limited universality of the research conclusions. It is worth noting that although Hyp has shown remarkable therapeutic effects in vitro, several in vivo model studies are required to facilitate its clinical application. Conclusion In this study, we identified key targets and signaling pathways of Hyp in the treatment of NSCLC using a network pharmacology method. In addition, our in vitro experiments confirmed that Hyp inhibited the proliferation and induced apoptosis of lung adenocarcinoma cells through regulation of the EGFR/ERK/FOXO1 pathway. These findings indicate that Hyp is a promising and effective multitarget drug. Our findings also emphasize the critical role of network pharmacology in target screening and pathway prediction for drug development. However, the follow-up work needs to be extended to squamous cell carcinoma cell lines, such as H1703 and SK-MES-1, to verify the universality of this mechanism in different histological subtypes. Supplementary Information Below is the link to the electronic supplementary material. [152]Supplementary Material 1.^ (11.7KB, xlsx) [153]Supplementary Material 2.^ (13KB, xlsx) [154]Supplementary Material 3.^ (11.3KB, xlsx) [155]Supplementary Material 4.^ (13.7KB, xlsx) [156]Supplementary Material 5.^ (10.4KB, xlsx) [157]Supplementary Material 6.^ (254.2KB, pdf) [158]Supplementary Material 7.^ (373.3KB, pdf) Acknowledgements