Skip to main content
Medicine logoLink to Medicine
. 2023 Dec 22;102(51):e36656. doi: 10.1097/MD.0000000000036656

Exploration of the effect and mechanism of Scutellaria barbata D. Don in the treatment of ovarian cancer based on network pharmacology and in vitro experimental verification

Jie Zhang a,*, Cong Qi b, He Li c, Chenhuan Ding c, Libo Wang a, Hongjin Wu a, Weiwei Dai a, Chenglong Wang a
PMCID: PMC10735072  PMID: 38134066

Abstract

The mortality rate of ovarian cancer is the highest among gynecological cancers, posing a serious threat to women health and life. Scutellaria barbata D. Don (SBD) can effectively treat ovarian cancer. However, its mechanism of action is unclear. The aim of this study was to elucidate the mechanism of SBD in the treatment of ovarian cancer using network pharmacology, and to verify the experimental results using human ovarian cancer SKOV3 cells. The Herb and Disease Gene databases were searched to identify common targets of SBD and ovarian cancer. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and Protein-Protein Interaction (PPI) network analyses were performed to identify the potential molecular mechanisms behind SBD. Finally, the molecular docking and main possible pathways were verified by experimental studies. Cell proliferation, the mRNA expression level of key genes and signaling pathway were all investigated and evaluated in vitro. A total of 29 bioactive ingredients and 137 common targets in SBD were found to inhibit ovarian cancer development. The active ingredients identified include quercetin, luteolin, and wogonin. Analysis of the PPI network showed that AKT1, VEGFA, JUN, TNF, and Caspase-3 shared centrality among all target genes. The results of the KEGG pathway analysis indicated that the cancer pathway, PI3K-Akt signaling pathway, and MAPK signaling pathways mediated the effects of SBD against ovarian cancer progression. Cell experiments showed that quercetin, luteolin, and wogonin inhibited the proliferation and clone formation of SKOV3 cells and regulated mRNA expression of 5 key genes by inhibiting PI3K/Akt signaling pathway. Our results demonstrate that SBD exerted anti-ovarian cancer effects through its key components quercetin, luteolin and wogonin. Mechanistically, its anti-cancer effects were mediated by inhibition of the PI3K/Akt signaling pathways. Therefore, SBD might be a candidate drug for ovarian cancer treatment.

Keywords: Scutellaria barbata D. Don, ovarian cancer, network pharmacology, molecular docking, experiments

1. Introduction

Ovarian cancer (OC) is a lethal and common gynecological malignancy, with 80% of patients diagnosed at an advanced stage of disease.[1] Fewer than half of OC patients survive beyond 5 years after diagnosis due to the prevalence of aggressive high-grade serous carcinomas and the lack of accurate early detection methods.[2] Despite many improvements in ovarian cancer diagnosis and treatment, conventional chemotherapy and new biological drugs cannot cure the disease, and the overall prognosis of patients remains poor.[3] Thus, traditional Chinese medicine (TCM), a complementary and alternative approach, is considered to be an effective alternative treatment for ovarian cancer. Some Chinese medicine preparations are widely used to treat cancer due to their properties of clearing away heat and toxic materials, and improving immunity thereby enhancing the quality of life of patients.[4]

The use of TCM in treating cancer has been recorded in Chinese medical texts and publications for more than 2 millennia. Scutellaria barbata D. Don (SBD) is derived from the dried Labiate plant and has been widely used to treat patients with multiple cancers.[5] SBD, belonging to Labiatae, has a sour taste and is cold in nature.[6] They have the functions of clearing away heat and detoxification, promoting blood circulation, removing blood stasis, anti-inflammatory and analgesic effects, and can be used as an adjuvant treatment for various cancers, including colorectal cancer, breast cancer, bladder cancer, lung cancer, liver cancer, gastric cancer, ovarian cancer, and other malignant tumors.[7,8] Studies have also shown that SBD can inhibit proliferation and induce apoptosis of ovarian cancer cell lines[9] and downregulate MMP-2/9 expression to inhibit the migration of ovarian cancer cells.[10] However, there are limited research related to SBD in the field of ovarian cancer, and most of them focus on a single target or a single pathway, which cannot comprehensively explain the antitumor effects of SBD.SBD contain multiple components and regulates several target genes with synergistic effects among various members. Therefore, the detailed effects and mechanism of SBD in the treatment of ovarian cancer need to be explored.

Network pharmacology combines bioinformatics, system biology, and pharmacology to explain the interaction between TCM and diseases and conforms to the systematic and holistic view of TCM.[11,12] Network pharmacology updates the “one target, one drug” model to the “multicomponent, multitarget” model. In addition, network pharmacology clarifies the complex interactions between genes, proteins, and metabolites related to diseases and drugs, allowing systematic analysis of the relationship between TCM and diseases.[13,14] This study combined network pharmacology with experimental verification to clarify the characteristics and potential molecular mechanism of SBD in treating ovarian cancer, opening up a new avenue for investigation. This research on SBD molecular mechanisms in ovarian cancer treatment not only lays a foundation for its broader use but also provides a reference for developing TCM and monomers, facilitating its integration with modern medicine to benefit patients, as shown in Figure 1, the working flowchart.

Figure 1.

Figure 1.

Flow diagram of the research.

2. Materials and methods

2.1. Network pharmacology analysis

2.1.1. Identification of active ingredients of SBD.

This study searched for the active ingredients of SBD in the Traditional Chinese Medicine System Pharmacology (TCMSP, https://old.tcmsp-e.com/index.php) database. The TCMSP database contains comprehensive information about traditional Chinese herbs, including their chemical structure, oral bioavailability, drug-likeness, half-life, Caco-2 intestinal epithelial permeability, blood-brain barrier, and associated drug-target-disease network. Subsequently, pharmacokinetic properties, including absorption, distribution, metabolism, and excretion (ADME), oral bioavailability (OB) ≥ 30%, and drug-likeness (DL) ≥ 0.18%, were used to screen for active ingredients of SBD. Compounds with OB ≥ 30% have good absorption, and a DL ≥ 0. 18% is chemically conducive to drug development.[15] We then obtained the SDF structural formulae of active ingredients from the PubChem database (http://pubchem.ncbi.nlm.nih.gov/) for ulterior target prediction.[16]

2.1.2. Prediction of potential targets of SBD.

This study used the TCMSP target module, SwissTargetPrediction, (http://www.swisstargetprediction.ch/), and PharmMapper (http://www.lilab-ecust.cn/pharmmapper/) databases to predict the potential targets of SBD active ingredients. The TCMSP database provides the chemical composition of Chinese herbs, all-sided targets of active ingredients, and their associated with diseases.[15] SwissTargetPrediction predicts the targets of active ingredients by comparing their structures to the 2- and 3-dimensional structures of known compounds.[17] PharmMapper is a freely accessible server that uses pharmacodynamic gene mapping methods to recognize candidate targets for drugs, natural products, or other newly found compounds with unidentified binding targets.[18] The resulting target protein names were ID mapped to their gene names using the UniProt database. (http://www.UniProt.org/).[19] All the targets were limited to “homo sapiens.”

2.1.3. Target prediction of ovarian cancer disease.

Four disease databases, including the Drugbank database (https://www.drugbank.ca/),[20] DisGeNET database (https://www.disgenet.org/home/),[21] GeneCards database (https://www.genecards.org/),[22] and OMIM database (https://omim.org/),[23] were integrated to obtain disease-related targets for ovarian cancer with “ovarian cancer” as the keyword. All the targets were limited to “homo sapiens.” The results retrieved at this stage were merged, and only one repeated target was reserved.

2.1.4. Intersection active ingredients and disease targets construction.

The Jvenn online tool was used to generate a Venn diagram and identify common targets for SBD and ovarian cancer. The identified common targets were then submitted to String (https://string-db.org/) to generate a protein-protein interaction (PPI) network.[24,25] The obtained network was visualized using Cytoscape v3.9.0, and topological analysis was performed using the network analysis module.

2.1.5. Biological function analysis of GO and KEGG pathway enrichment analysis.

To gain insight into the gene functions and signaling pathways of the SBD targets, we used the Metascape (https://www.metascape.org/) tools,[26,27] which are online biological knowledge base and analysis tools, to identify enriched cellular components (CC), biological processes (BP), molecular function (MF), and KEGG pathways. GO analysis is mainly used to describe the functions of gene products, including cellular, molecular, and biological functions. On the other hand, KEGG pathway enrichment analysis is used to analyze core pathways and explore the possible biological function and signaling pathway mechanisms in the treatment of ovarian cancer. The GO and KEGG results were visually analyzed by on the online bioinformatics platform (http://www.bioinformatics.com.cn/) and with the Cytoscape software.

2.2. Molecular docking analysis

The 2D structures of the potential active ingredients in SBD were downloaded from the TCMSP database while the 3D structure of the ovarian cancer docking targets (top 5 of degree in PPI network) treated by SBD was downloaded from the Worldwide Protein Data Bank (PDB) database (https://www.rcsb.org/).[28] They were imported to AutoDockTools[29] for hydrogenation, dehydration, and other pretreatments. Then, molecular docking of the receptor and ligand was conducted to analyze its binding activity. The docking results were visualized using the PyMol software.[30]

2.3. Experimental verification

2.3.1. Cell culture, drugs, and reagents.

The SKOV3 cells were maintained in DMEM (Gibco, USA) with 10% FBS (Gibco, USA), 100 U/mL penicillin, and 100 g/mL streptomycin (Sigma, USA). The cells were cultured at 37°C in a water-saturated atmosphere under 5% CO2.

Quercetin (Sigma, USA, purity ≥ 99.0%) and luteolin and wogonin (Shanghai Yuanye, China, purity ≥ 98.0%) were dissolved in dimethyl sulfoxide (DMSO)The final amount of DMSO added to the cell culture medium was <0.1% to ensure that there were no effects on cells.

MTT Kit (CCK) − 8 (Beyotime, China), paraformaldehyde (Beyotime, China), crystal violet (Sigma, USA), TRIzol solution (Life, China), ReverTra Ace Qpcr RT Kit (Takara, Japan), SYBR-Green Real-Time PCR Master Mix kit (Takara, Japan), Primers (Sangon Biotech, China), BCA protein Quantifier kit (Invitrogen, USA), and anti-PI3K, anti-AKT, anti-p-AKT, anti-Caspase (CST, USA) were used in this study.

2.3.2. Cell proliferation assay.

The MTT assay kit was used to assess cell proliferation. The SKOV3 cells at the logarithmic growth stage were digested with 0.25% trypsin and cultured in a 96-well plate at a density of 4 × 103/well and a volume of 100 µL per well for 24 hours. This study had 5 replicate wells in each experimental group. After incubation, different concentrations of quercetin, luteolin, and wogonin (0, 30 µM, 60 µM, 90 µM, 120 µM, and 150 µM) were added to the culture. The cells were then cultured for 24 and 48 hours to observe the cell morphology. Then, 20 μL MTT solution was added to each well and incubated in the dark for 4 hours. Subsequently, the supernatant was discarded, and 150 μL DMSO was added to each well to dissolve methylzan crystals. Finally, the absorbance (OD) of the experimental wells was measured at 490 nm using a microplate reader. The Inhibition Rate (IR) of cell proliferation was calculated using the formula: IR % =1   OD   value   of   drug   action   groupOD value of control group×100%. Half maximal inhibitory concentration(IC50) was calculated and was used for subsequent experiments.

2.3.3. Colony formation assay.

SKOV3 cells in the logarithmic growth stage were digested with 0.25% trypsin and inoculated in a 6 cm dish (1200 cells/well) to perform a colony formation assay. Then, the IC50 concentrations of quercetin, luteolin, and wogonin were added to the culture and incubated for 48 hours. The drug-containing medium was discarded and washed with PBS, and a fresh medium containing 10% FBS was added. The medium was changed every 3 days, and the culture was incubated for 8 days. The assay was stopped once the colonies were visible to the naked eye. The original culture medium was discarded, and the cells were carefully washed twice with precooled PBS. The cells were then fixed with 4% paraformaldehyde for 30 minutes. The colonies were stained with 0.1% crystal violet for 20 minutes at 37 °C, and colonies containing more than 50 cells were counted using microscopy. Finally, the colony formation rate was calculated using the formula: Colony   formation   rate=number of cell coloniesnumber of inoculated cells×100%.

2.3.4. Reverse transcription quantitative polymerase chain reaction (RT-qPCR).

After 48 hours of quercetin, luteolin, and wogonin intervention, SKOV3 cells were washed twice with precooled PBS. TRIzol solution was added to extract the total RNA of the SKOV3 cells. The PrimeScript RT reagent Kit was used to reverse RNA transcription into cDNA. The designed cDNA was used in the amplification process using TB Green® Premix Ex Taq. The accumulation of PCR products was monitored in real-time, and simultaneous determination of cycle threshold (Ct) values was conducted. Gene expression values were determined using the 2−ΔΔCt method. The primer designs of the hub genes are shown in Table 1.

Table 1.

Primer sequences of PCR.

Gene Primer sequence(5‘-3‘) Length/bp
18s F: CGTTGATTAAGTCCCTGCCCTT 137
R: TCAAGTTCGACCGTCTTCTCAG
AKT1 F: AGCGACGTGGCTATTGTGAAG 96
R: GCCATCATTCTTGAGGAGGAAGT
VEGFA F: AGGGCAGAATCATCACGAAGT 75
R: AGGGTCTCGATTGGATGGCA
Caspase 3 F: GAAATTGTGGAATTGATGCGTGA 164
R: CTACAACGATCCCCTCTGAAAAA
Jun F: TCCAAGTGCCGAAAAAGGAAG 78
R: CGAGTTCTGAGCTTTCAAGGT
TNF F: GAGGCCAAGCCCTGGTATG 91
R: CGGGCCGATTGATCTCAGC

2.3.5. Western blotting analysis.

After 48 hours of quercetin, luteolin, and wogonin intervention, the protein lysate was added to the ice bath. The total protein concentration was determined quantitatively using the BCA protein concentration assay kit. After electrophoresis with 10% SDS-PAGE, the separated proteins from the gel were transferred onto the PVDF membrane, and 5% BSA blocking buffer was added and incubated for 1 hour. The primary antibodies against PI3K, Akt, P-Akt, Caspase-3, and GAPDH were used according to the manufactures’ instructions and incubated at 4 °C overnight. Next, the membrane was washed with TBST 5 times for 8 minutes each time, and then incubated with the corresponding secondary antibody (1:3000) at room temperature for 1 hour and then washed with TBST 5 times for 8 minutes each time. A ECL luminescence kit was used for color development, and exposed to the gel imaging system. Image J software was used to analyze protein bands and calculate gray values. The gray ratio of target protein/GAPDH protein represented its relative expression level.

2.4. Statistical analysis

The data were processed using SPSS 22.0 statistical software and presented as the mean ± standard deviation if they obeyed normal distribution. One-way ANOVA was used to compare the measurement data between single-factor design and multiple groups. The least significant difference method was used for multiple comparisons, and the Games–Howell test was used to test the variance. The results analysis map was prepared using GraphPad Prism 8.0 software.

3. Results

3.1. Network pharmacology analysis

3.1.1. Screening of active ingredients.

A total of 94 known active compounds of SBD, shown in Supplementary Table S1, http://links.lww.com/MD/L93, were identified from the TCMSP database. These active compounds were screened based on OB ≥ 30% and DL ≥ 0.18, yielding 29 active ingredients which met the conditions. The results are presented in Table 2.

Table 2.

Basic information on the main active ingredients of SBD.

Number MOL ID Active ingredients OB (%) DL
SBD1 MOL001040 (2R)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 42.36 0.21
SBD2 MOL012245 5,7,4’-trihydroxy-6-methoxyflavanone 36.63 0.27
SBD3 MOL012246 5,7,4’-trihydroxy-8-methoxyflavanone 74.24 0.26
SBD4 MOL012248 5-hydroxy-7,8-dimethoxy-2-(4-methoxyphenyl)chromone 65.82 0.33
SBD5 MOL012250 7-hydroxy-5,8-dimethoxy-2-phenyl-chromone 43.72 0.25
SBD6 MOL012251 Chrysin-5-methylether 37.27 0.2
SBD7 MOL012252 9,19-cyclolanost-24-en-3-ol 38.69 0.78
SBD8 MOL002776 Baicalin 40.12 0.75
SBD9 MOL012254 campesterol 37.58 0.71
SBD10 MOL000953 CLR 37.87 0.68
SBD11 MOL000358 beta-sitosterol 36.91 0.75
SBD12 MOL012266 rivularin 37.94 0.37
SBD13 MOL001973 Sitosteryl acetate 40.39 0.85
SBD14 MOL012269 Stigmasta-5,22-dien-3-ol-acetate 46.44 0.86
SBD15 MOL012270 Stigmastan-3,5,22-triene 45.03 0.71
SBD16 MOL000449 Stigmasterol 43.83 0.76
SBD17 MOL000173 wogonin 30.68 0.23
SBD18 MOL001735 Dinatin 30.97 0.27
SBD19 MOL001755 24-Ethylcholest-4-en-3-one 36.08 0.76
SBD20 MOL002714 baicalein 33.52 0.21
SBD21 MOL002719 6-Hydroxynaringenin 33.23 0.24
SBD22 MOL002915 Salvigenin 49.07 0.33
SBD23 MOL000351 Rhamnazin 47.14 0.34
SBD24 MOL000359 sitosterol 36.91 0.75
SBD25 MOL005190 eriodictyol 71.79 0.24
SBD26 MOL005869 daucostero_qt 36.91 0.75
SBD27 MOL000006 luteolin 36.16 0.25
SBD28 MOL008206 Moslosooflavone 44.09 0.25
SBD29 MOL000098 quercetin 46.43 0.28

3.1.2. Potential targets of active ingredients.

Candidate targets of active components in SBD were searched on the TCMSP and,SwissTarget Prediction and PharmMapper databases. UniProt was used to standardize the nomenclature for the active compounds. After UniProt standardization, deduplication occurred. Duplicate data were removed, leaving 229 targets, shown in Supplementary Table S2, http://links.lww.com/MD/L94. The CytoScape 3.9.0 software was then used to build an “active ingredients–targets” interaction network, as shown in Figure 2. In the study, a total of 29 active components of SBD were identified. According to the degree value of the interaction between the active component and the target, 3 core active components (quercetin, luteolin, wogonin) were identified.

Figure 2.

Figure 2.

“Ingredients–targets” network construction. The light orange prism nodes represent the targets, and the light blue round nodes represent SBD ingredients. SBD = Scutellaria barbata D. Don.

3.1.3. Potential targets of disease.

The GeneCards, DisGeNet, OMIM, and Drugbank databases were used to identify reviewed therapeutic targets in SBD. The scores in GeneCards and DisGeNet were used to define a set of high-confidence interactions between diseases and potential targets, with a relevance score ≥ 12.7 in GeneCards and a score-GDA ≥ 0.1 in DisGeNet. As a result, 1141 ovarian cancer-related targets were acquired from GeneCards, 1224 ovarian cancer-related targets were obtained from DisGeNet, 44 ovarian cancer-related targets were obtained from OMIM, and 75 ovarian cancer-related targets were obtained from Drugbank. After removing the duplicate targets, 1969 targets for ovarian cancer remained, as shown in Supplementary Table S3, http://links.lww.com/MD/L95.

3.1.4. Drug–disease intersection targets.

Venn analysis was performed using the 229 targets of SBD active components and 1969 ovarian cancer-related target genes, and 137 drug–disease intersection gene targets were obtained for further analysis, as shown in Figure 3 and Table 3.

Figure 3.

Figure 3.

(A) Venn diagram shows ovarian cancer-related targets among the 4 databases. (B) Venn diagram of common intersecting targets genes in SBD and Ovarian cancer. SBD = Scutellaria barbata D. Don.

Table 3.

Potential targets of SBD against ovarian cancer.

Serial number Gene
name
Protein
name
Uniprot
ID
1 PTGS1 Prostaglandin G/H synthase 1 P23219
2 ESR1 Estrogen receptor P03372
3 PTGS2 Prostaglandin G/H synthase 2 P35354
4 PRKACA cAMP-dependent protein kinase catalytic subunit alpha P17612
5 PGR Progesterone receptor P06401
6 NR3C1 Glucocorticoid receptor P04150
7 PIK3CG Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform P48736
8 NOS2 Nitric oxide synthase, inducible P35228
9 KCNH2 Potassium voltage-gated channel subfamily H member 2 Q12809
10 AR Androgen receptor P10275
11 F10 Coagulation factor X P00742
12 NOS3 Nitric oxide synthase, endothelial P29474
13 ESR2 Estrogen receptor beta Q92731
14 GSK3B Glycogen synthase kinase-3 beta P49841
15 CDK2 Cyclin-dependent kinase 2 P24941
16 CHEK1 Serine/threonine-protein kinase Chk1 O14757
17 PRSS1 Trypsin-1 P07477
18 PPARG Peroxisome proliferator-activated receptor gamma P37231
19 ADRA1D Alpha-1D adrenergic receptor P25100
20 MAPK14 Mitogen-activated protein kinase 14 Q16539
21 ADRA1A Alpha-1A adrenergic receptor P35348
22 PTPN1 Tyrosine-protein phosphatase non-receptor type 1 P18031
23 BCL2 Apoptosis regulator Bcl-2 P10415
24 BAX Apoptosis regulator BAX Q07812
25 CASP9 Caspase-9 P55211
26 JUN Transcription factor AP-1 P05412
27 CASP3 Caspase-3 P42574
28 CASP8 Caspase-8 Q14790
29 PRKCA Protein kinase C alpha type P17252
30 TGFB1 Transforming growth factor beta-1 proprotein P01137
31 PON1 Serum paraoxonase/arylesterase 1 P27169
32 MAP2 Microtubule-associated protein 2 P11137
33 KDR Vascular endothelial growth factor receptor 2 P35968
34 PLAU Urokinase-type plasminogen activator P00749
35 RELA Transcription factor p65 Q04206
36 AKT1 RAC-alpha serine/threonine-protein kinase P31749
37 CCND1 G1/S-specific cyclin-D1 P24385
38 CDKN1A Cyclin-dependent kinase inhibitor 1 P38936
39 EIF6 Eukaryotic translation initiation factor 6 P56537
40 TNF Tumor necrosis factor P01375
41 IL6 Interleukin-6 P05231
42 AHSA1 Activator of 90 kDa heat shock protein ATPase homolog 1 O95433
43 TP53 Cellular tumor antigen p53 P04637
44 MMP1 Interstitial collagenase P03956
45 CCL2 C-C motif chemokine 2 P13500
46 PRKCD Protein kinase C delta type Q05655
47 FN1 Fibronectin P02751
48 CXCL8 Interleukin-8 P10145
49 MCL1 Induced myeloid leukemia cell differentiation protein Mcl-1 Q07820
50 VEGFA Vascular endothelial growth factor A P15692
51 FOS Proto-oncogene c-Fos P01100
52 MMP9 Matrix metalloproteinase-9 P14780
53 HIF1A Hypoxia-inducible factor 1-alpha Q16665
54 FOSL1 Fos-related antigen 1 P15407
55 CDK1 Cyclin-dependent kinase 1 P06493
56 CCNB1 G2/mitotic-specific cyclin-B1 P14635
57 MPO Myeloperoxidase P05164
58 AHR Aryl hydrocarbon receptor P35869
59 IGF2 Insulin-like growth factor II P01344
60 CYCS Cytochrome c P99999
61 ALOX12 Polyunsaturated fatty acid lipoxygenase ALOX12 P18054
62 NFATC1 Nuclear factor of activated T-cells, cytoplasmic 1 O95644
63 CCNA2 Cyclin-A2 P20248
64 HMOX1 Heme oxygenase 1 P09601
65 NFE2L2 Nuclear factor erythroid 2-related factor 2 Q16236
66 NQO1 NAD P15559
67 EGFR Epidermal growth factor receptor P00533
68 BCL2L1 Bcl-2-like protein 1 Q07817
69 MMP2 72 kDa type IV collagenase P08253
70 MAPK1 Mitogen-activated protein kinase 1 P28482
71 IL10 Interleukin-10 P22301
72 RB1 Retinoblastoma-associated protein P06400
73 CDK4 Cyclin-dependent kinase 4 P11802
74 NFKBIA NF-kappa-B inhibitor alpha P25963
75 TOP1 DNA topoisomerase 1 P11387
76 MDM2 E3 ubiquitin-protein ligase Mdm2 Q00987
77 PCNA Proliferating cell nuclear antigen P12004
78 ERBB2 Receptor tyrosine-protein kinase erbB-2 P04626
79 CASP7 Caspase-7 P55210
80 ICAM1 Intercellular adhesion molecule 1 P05362
81 BIRC5 Baculoviral IAP repeat-containing protein 5 O15392
82 IL2 Interleukin-2 P60568
83 TYR Tyrosinase P14679
84 IFNG Interferon gamma P01579
85 IL4 Interleukin-4 P05112
86 TOP2A DNA topoisomerase 2-alpha P11388
87 GSTP1 Glutathione S-transferase P P09211
88 XIAP E3 ubiquitin-protein ligase XIAP P98170
89 INSR Insulin receptor P06213
90 CD40LG CD40 ligand P29965
91 MET Hepatocyte growth factor receptor P08581
92 MMP3 Stromelysin-1 P08254
93 EGF Pro-epidermal growth factor P01133
94 CDKN2A Cyclin-dependent kinase inhibitor 2A P42771
95 ELK1 ETS domain-containing protein Elk-1 P19419
96 ODC1 Ornithine decarboxylase P11926
97 RAF1 RAF proto-oncogene serine/threonine-protein kinase P04049
98 SOD1 Superoxide dismutase P00441
99 STAT1 Signal transducer and activator of transcription 1-alpha/beta P42224
100 HSPA5 Endoplasmic reticulum chaperone BiP P11021
101 CYP3A4 Cytochrome P450 3A4 P08684
102 CYP1A2 Cytochrome P450 1A2 P05177
103 CAV1 Caveolin-1 Q03135
104 MYC Myc proto-oncogene protein P01106
105 F3 Tissue factor P13726
106 GJA1 Gap junction alpha-1 protein P17302
107 CYP1A1 Cytochrome P450 1A1 P04798
108 IL1B Interleukin-1 beta P01584
109 VCAM1 Vascular cell adhesion protein 1 P19320
110 HSPB1 Heat shock protein beta-1 P04792
111 SULT1E1 Sulfotransferase 1E1 P49888
112 NR1I2 Nuclear receptor subfamily 1 group I member 2 O75469
113 CYP1B1 Cytochrome P450 1B1 Q16678
114 THBD Thrombomodulin P07204
115 SERPINE1 Plasminogen activator inhibitor 1 P05121
116 ALOX5 Polyunsaturated fatty acid 5-lipoxygenase P09917
117 PTEN Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN P60484
118 IL1A Interleukin-1 alpha P01583
119 ABCA2 ATP-binding cassette sub-family A member 2 Q9BZC7
120 PARP1 Poly P09874
121 CXCL11 C-X-C motif chemokine 11 O14625
122 CHEK2 Serine/threonine-protein kinase Chk2 O96017
123 CLDN4 Claudin-4 O14493
124 HSF1 Heat shock factor protein 1 Q00613
125 CRP C-reactive protein P02741
126 CHUK Inhibitor of nuclear factor kappa-B kinase subunit alpha O15111
127 SPP1 Osteopontin P10451
128 RUNX2 Runt-related transcription factor 2 Q13950
129 RASSF1 Ras association domain-containing protein 1 Q9NS23
130 E2F1 Transcription factor E2F1 Q01094
131 E2F2 Transcription factor E2F2 Q14209
132 CTSD Cathepsin D P07339
133 IGFBP3 Insulin-like growth factor-binding protein 3 P17936
134 IRF1 Interferon regulatory factor 1 P10914
135 ERBB3 Receptor tyrosine-protein kinase erbB-3 P21860
136 HK2 Hexokinase-2 P52789
137 GSTM1 Glutathione S-transferase Mu 1 P09488

3.1.5. PPI network analysis.

The String database was used to analyze the interactions between the 137 shared targets. The String analysis contained 137 nodes, 3071 link edges, and 956 expected edges at the medium confidence. The average node degree was 44.8, the average local clustering coefficient was 0.706, and the PPI enrichment P value was < 0.001. Afterward, the Cytoscape software was used to construct the PPI network. Different colors and rhombus sizes represent different biological information. The larger circles with darker colors (from orange to red) indicate stronger interactions, as shown in Figure 4A. In addition, a topological analysis of the 137 common targets was performed using a Cytoscape analysis module, shown in Table 4. The top 50 targets were ranked according to their degree values, represented by a bar graph, shown in Figure 4B.

Figure 4.

Figure 4.

PPI network of 137 target genes and top 50 targets from the PPI network. (A) Visualization of the PPI network of 137 target genes: from orange to red, the larger rhombus suggests stronger interactions, and the central region is the top 50 targets. (B) A histogram of the top 50 targets from the PPI network. The numbers represent degree values. PPI = protein-protein interaction.

Table 4.

The topological analysis of top 50 genes. Data were ranked by degree.

Gene name Degree BetweennessCentrality ClosenessCentrality TopologicalCoefficient
TP53 115 0.043158555 0.88590604 0.383662714
AKT1 111 0.04562599 0.862745098 0.386022386
VEGFA 102 0.024739627 0.814814815 0.409907903
JUN 102 0.023990559 0.814814815 0.413770053
TNF 99 0.026484946 0.8 0.408478727
ESR1 99 0.038618348 0.8 0.398454239
MYC 98 0.020122447 0.795180723 0.412569573
IL6 96 0.022084432 0.785714286 0.41635101
CASP3 96 0.018645246 0.785714286 0.426136364
EGFR 93 0.025142474 0.771929825 0.417481264
HIF1A 93 0.01727923 0.771929825 0.430188987
EGF 88 0.017439426 0.75 0.434831267
PTGS2 88 0.024499214 0.75 0.431215565
CCND1 88 0.01359779 0.75 0.432162534
IL1B 87 0.012254171 0.745762712 0.430947405
PTEN 86 0.013018932 0.741573034 0.431994362
MMP9 85 0.014327873 0.737430168 0.442869875
ERBB2 80 0.017517308 0.717391304 0.435132576
FOS 79 0.010367113 0.709677419 0.458691661
PPARG 76 0.010672586 0.70212766 0.447468102
FN1 75 0.006577537 0.698412698 0.464242424
CXCL8 74 0.007114304 0.694736842 0.457002457
BCL2L1 74 0.006162498 0.691099476 0.471322468
CASP8 74 0.00737131 0.694736842 0.463042588
NFKBIA 72 0.005065923 0.683937824 0.479961832
CDKN2A 70 0.011972217 0.676923077 0.460196292
MAPK1 68 0.009270717 0.673469388 0.448975045
CYCS 68 0.009060935 0.673469388 0.467134581
IL10 66 0.004331235 0.663316583 0.480222068
MMP2 66 0.003485736 0.666666667 0.4890955
CCL2 66 0.005349782 0.666666667 0.46751607
RELA 63 0.003272746 0.653465347 0.495456198
MDM2 63 0.005222524 0.656716418 0.456108706
CASP9 62 0.002972236 0.650246305 0.497168185
CDKN1A 62 0.003743578 0.650246305 0.491135188
HMOX1 61 0.005224187 0.647058824 0.468777375
STAT1 61 0.003952042 0.647058824 0.49518208
MAPK14 60 0.004906218 0.643902439 0.49389313
ICAM1 58 0.002734795 0.637681159 0.484601211
GSK3B 57 0.004944944 0.637681159 0.487107921
IL2 56 0.002933688 0.628571429 0.509478022
IL4 56 0.002752027 0.631578947 0.496864776
AR 56 0.004264953 0.634615385 0.4745671
NOS3 56 0.004161875 0.634615385 0.472267316
MCL1 56 0.002805555 0.634615385 0.501217532
CAV1 55 0.010162808 0.631578947 0.484848485
CDK4 55 0.005392789 0.625592417 0.472867133
TGFB1 55 0.001273775 0.631578947 0.518595041
IFNG 54 0.00179449 0.625592417 0.508340401
PARP1 53 0.003632184 0.61971831 0.478664731

3.1.6. GO/KEGG function and pathway enrichment analysis.

The Metascape data platform was used for enrichment analysis of the 137 relevant ovarian cancer-related targets of SBD. The results were visualized using online biological tools. GO and KEGG pathways with FDR < 0.01 were considered significantly enriched. The GO and KEGG enrichment analyses were performed on the 137 therapeutic targets to obtain 2185 pathways (P < .01). In total, 1766, 81, 145, and 193 pathways were identified in GOBP, GOCC, GOMF, and KEGG, respectively (all P < .01), as shown in Supplementary Table S4, http://links.lww.com/MD/L96. The top 20 enriched GO terms are listed in Figure 5. The biological process involves responses to inorganic substances, xenobiotic stimuli, hormones, reactive oxygen species, positive cellular component movement regulation, and positive cell death regulation. On the other hand, CC involves a transcription regulator complex, membrane raft, membrane microdomain, cyclin-dependent protein kinase holoenzyme complex, vesicle lumen, and outer organelle membrane, and MF involves kinase binding, DNA-binding transcription factor binding, transcription factor binding, protein kinase activity, protein domain-specific binding, and cytokine receptor binding.

Figure 5.

Figure 5.

GO enrichment analysis of the 137 potential therapeutic targets. (A) Dot plot of GO enrichment analysis: the top 20 BP terms. (B) GO enrichment analysis: the top 20 MF terms. (C) GO enrichment analysis: the top 20 CC terms. (A–C) Node color is displayed in a gradient from red to green in descending order of the P value. The size of the nodes is arranged in ascending order of the number of genes. (D) Enrichment of GO Term (the top ten) BP, CC, MF 3 in one histogram. BP = biological processes, CC = cellular components, GO = Gene Ontology, MF = molecular function.

The top 20 noticeably enriched KEGG terms related to cancer are listed in Figure 6. The results of the KEGG enrichment analysis showed that the cancer pathway and PI3K-Akt signaling pathway are the main anti-ovarian cancer pathways of SBD. The KEGG Mapper platform was used to draw the pathway mechanism diagram of the 2 signaling pathways. The specific mechanism of SBD in treating ovarian cancer was further analyzed according to the pathway mechanism diagram.

Figure 6.

Figure 6.

KEGG pathway enrichment analysis of the 137 potential therapeutic targets. (A) Bubble diagram of KEGG analysis. (B) Bar plot of KEGG analysis, sorted by the importance of–log10(P) of each lane. (C) Pathways in cancer. (d) PI3K-AKT signal pathway (red nodes represent 50 core targets in the pathway). KEGG = Kyoto Encyclopedia of Genes and Genomes.

Table 5 shows the main signaling pathways with enrichment targets ≥ 20. The pathways include the cancer pathway, PI3K-Akt signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, Proteoglycans in cancer, Chemical carcinogenesis- receptor activation, MicroRNAs in cancer, Apoptosis, p53 signaling pathway, TNF signaling pathway, IL-17 signaling pathway and so on.

Table 5.

The signaling pathway of KEGG enrichment target number ≥ 20.

GO Pathway Count Genes
hsa05200 Pathways in cancer 65 AKT1 XIAP BIRC5 AR BAX CCND1 BCL2 BCL2L1 CASP3 CASP7 CASP8 CASP9 CCNA2 CDK2 CDK4 CDKN1A CDKN2A CHUK NQO1 E2F1 E2F2 EGF EGFR ELK1 ERBB2 ESR1 ESR2 FN1 FOS GSK3B GSTM1 GSTP1 HIF1A HMOX1 IFNG IGF2 IL2 IL4 IL6 CXCL8 JUN MDM2 MET MMP1 MMP2 MMP9 MYC NFE2L2 NFKBIA NOS2 PPARG PRKACA PRKCA MAPK1 PTEN PTGS2 RAF1 RB1 RELA STAT1 TGFB1 TP53 VEGFA RASSF1 CYCS
hsa05417 Lipid and atherosclerosis 36 AKT1 BAX BCL2 BCL2L1 CASP3 CASP7 CASP8 CASP9 CD40LG CHUK MAPK14 CYP1A1 FOS GSK3B HSPA5 ICAM1 IL1B IL6 CXCL8 JUN MMP1 MMP3 MMP9 NFATC1 NFE2L2 NFKBIA NOS3 PPARG PRKCA MAPK1 RELA CCL2 TNF TP53 VCAM1 CYCS
hsa05163 Human cytomegalovirus infection 35 AKT1 BAX CCND1 CASP3 CASP8 CASP9 CDK4 CDKN1A CDKN2A CHUK MAPK14 E2F1 E2F2 EGFR ELK1 GSK3B IL1B IL6 CXCL8 MDM2 MYC NFATC1 NFKBIA PRKACA PRKCA MAPK1 PTGS2 RAF1 RB1 RELA CCL2 TNF TP53 VEGFA CYCS
hsa04151 PI3K-Akt signaling pathway 35 AKT1 CCND1 BCL2 BCL2L1 CASP9 CDK2 CDK4 CDKN1A CHUK EGF EGFR ERBB2 ERBB3 FN1 GSK3B IGF2 IL2 IL4 IL6 INSR KDR MCL1 MDM2 MET MYC NOS3 PIK3CG PRKCA MAPK1 PTEN RAF1 RELA SPP1 TP53 VEGFA
hsa05161 Hepatitis B 34 AKT1 BIRC5 BAX BCL2 CASP3 CASP8 CASP9 CCNA2 CDK2 CDKN1A CHUK MAPK14 E2F1 E2F2 ELK1 FOS IL6 CXCL8 JUN MMP9 MYC NFATC1 NFKBIA PCNA PRKCA MAPK1 RAF1 RB1 RELA STAT1 TGFB1 TNF TP53 CYCS
hsa05166 Human T-cell leukemia virus 1 infection 33 AKT1 XIAP BAX CCND1 BCL2L1 CCNA2 CDK2 CDK4 CDKN1A CDKN2A CHEK1 CHUK E2F1 E2F2 ELK1 FOS ICAM1 IL2 IL6 JUN MYC NFATC1 NFKBIA PRKACA MAPK1 PTEN RB1 RELA TGFB1 TNF TP53 FOSL1 CHEK2
hsa05167 Kaposi sarcoma-associated herpesvirus infection 32 AKT1 BAX CCND1 CASP3 CASP8 CASP9 CDK4 CDKN1A CHUK MAPK14 E2F1 E2F2 FOS GSK3B HIF1A ICAM1 IL6 CXCL8 JUN MYC NFATC1 NFKBIA PIK3CG MAPK1 PTGS2 RAF1 RB1 RELA STAT1 TP53 VEGFA CYCS
hsa04933 AGE-RAGE signaling pathway in diabetic complications 30 AKT1 BAX CCND1 BCL2 CASP3 CDK4 MAPK14 F3 FN1 ICAM1 IL1A IL1B IL6 CXCL8 JUN MMP2 NFATC1 NOS3 SERPINE1 PRKCA PRKCD MAPK1 RELA CCL2 STAT1 TGFB1 THBD TNF VCAM1 VEGFA
hsa04218 Cellular senescence 29 AKT1 CCND1 CCNA2 CCNB1 CDK1 CDK2 CDK4 CDKN1A CDKN2A CHEK1 MAPK14 E2F1 E2F2 IGFBP3 IL1A IL6 CXCL8 MDM2 MYC NFATC1 SERPINE1 MAPK1 PTEN RAF1 RB1 RELA TGFB1 TP53 CHEK2
hsa05205 Proteoglycans in cancer 29 AKT1 CCND1 CASP3 CAV1 CDKN1A MAPK14 EGFR ELK1 ERBB2 ERBB3 ESR1 FN1 HIF1A IGF2 KDR MDM2 MET MMP2 MMP9 MYC PLAU PRKACA PRKCA MAPK1 RAF1 TGFB1 TNF TP53 VEGFA
hsa04010 MAPK signaling pathway 29 AKT1 CASP3 CHUK MAPK14 EGF EGFR ELK1 ERBB2 ERBB3 FOS HSPB1 IGF2 IL1A IL1B INSR JUN KDR MET MYC NFATC1 PRKACA PRKCA MAPK1 RAF1 RELA TGFB1 TNF TP53 VEGFA
hsa05165 Human papillomavirus infection 29 AKT1 BAX CCND1 CASP3 CASP8 CCNA2 CDK2 CDK4 CDKN1A CHUK E2F1 EGF EGFR FN1 GSK3B IRF1 MDM2 PRKACA MAPK1 PTEN PTGS2 RAF1 RB1 RELA SPP1 STAT1 TNF TP53 VEGFA
hsa05207 Chemical carcinogenesis - receptor activation 28 AHR AKT1 XIAP BIRC5 AR CCND1 BCL2 CYP1A1 CYP1A2 CYP1B1 CYP3A4 E2F1 EGF EGFR ESR1 ESR2 FOS GSTM1 JUN MYC PGR PRKACA PRKCA MAPK1 RAF1 RB1 RELA VEGFA
hsa05418 Fluid shear stress and atherosclerosis 27 AKT1 BCL2 CAV1 CHUK MAPK14 NQO1 FOS GSTM1 GSTP1 HMOX1 ICAM1 IFNG IL1A IL1B JUN KDR MMP2 MMP9 NFE2L2 NOS3 RELA CCL2 THBD TNF TP53 VCAM1 VEGFA
hsa05160 Hepatitis C 27 AKT1 BAX CCND1 CASP3 CASP8 CASP9 CDK2 CDK4 CDKN1A CHUK CLDN4 E2F1 E2F2 EGF EGFR GSK3B IFNG MYC NFKBIA MAPK1 RAF1 RB1 RELA STAT1 TNF TP53 CYCS
hsa05225 Hepatocellular carcinoma 27 AKT1 BAX CCND1 BCL2L1 CDK4 CDKN1A CDKN2A NQO1 E2F1 E2F2 EGFR ELK1 GSK3B GSTM1 GSTP1 HMOX1 IGF2 MET MYC NFE2L2 PRKCA MAPK1 PTEN RAF1 RB1 TGFB1 TP53
hsa05169 Epstein-Barr virus infection 27 AKT1 BAX CCND1 BCL2 CASP3 CASP8 CASP9 CCNA2 CDK2 CDK4 CDKN1A CHUK MAPK14 E2F1 E2F2 ICAM1 IL6 JUN MDM2 MYC NFKBIA RB1 RELA STAT1 TNF TP53 CYCS
hsa05215 Prostate cancer 26 AKT1 AR CCND1 BCL2 CASP9 CDK2 CDKN1A CHUK E2F1 E2F2 EGF EGFR ERBB2 GSK3B GSTP1 MDM2 MMP3 MMP9 NFKBIA PLAU MAPK1 PTEN RAF1 RB1 RELA TP53
hsa05208 Chemical carcinogenesis - reactive oxygen species 26 AHR AKT1 CHUK MAPK14 CYP1A1 CYP1A2 CYP1B1 NQO1 EGF EGFR FOS GSTM1 HIF1A HMOX1 JUN MET NFE2L2 NFKBIA PRKCD MAPK1 PTEN PTPN1 RAF1 RELA SOD1 VEGFA
hsa05206 MicroRNAs in cancer 26 CCND1 BCL2 CASP3 CDKN1A CDKN2A CYP1B1 E2F1 E2F2 EGFR ERBB2 ERBB3 HMOX1 MCL1 MDM2 MET MMP9 MYC PLAU PRKCA MAPK1 PTEN PTGS2 RAF1 TP53 VEGFA RASSF1
hsa05222 Small cell lung cancer 24 AKT1 XIAP BAX CCND1 BCL2 BCL2L1 CASP3 CASP9 CDK2 CDK4 CDKN1A CHUK E2F1 E2F2 FN1 MYC NFKBIA NOS2 PTEN PTGS2 RB1 RELA TP53 CYCS
hsa01522 Endocrine resistance 24 AKT1 BAX CCND1 BCL2 CDK4 CDKN1A CDKN2A MAPK14 E2F1 E2F2 EGFR ERBB2 ESR1 ESR2 FOS JUN MDM2 MMP2 MMP9 PRKACA MAPK1 RAF1 RB1 TP53
hsa04210 Apoptosis 23 PARP1 AKT1 XIAP BIRC5 BAX BCL2 BCL2L1 CASP3 CASP7 CASP8 CASP9 CHUK CTSD FOS JUN MCL1 NFKBIA MAPK1 RAF1 RELA TNF TP53 CYCS
hsa05162 Measles 23 AKT1 BAX CCND1 BCL2 BCL2L1 CASP3 CASP8 CASP9 CDK2 CDK4 CHUK FOS GSK3B IL1A IL1B IL2 IL6 JUN NFKBIA RELA STAT1 TP53 CYCS
hsa05164 Influenza A 23 AKT1 BAX CASP3 CASP8 CASP9 CDK4 CHUK ICAM1 IFNG IL1A IL1B IL6 CXCL8 NFKBIA PRKCA MAPK1 PRSS1 RAF1 RELA CCL2 STAT1 TNF CYCS
hsa05212 Pancreatic cancer 22 AKT1 BAX CCND1 BCL2L1 CASP9 CDK4 CDKN1A CDKN2A CHUK E2F1 E2F2 EGF EGFR ERBB2 MAPK1 RAF1 RB1 RELA STAT1 TGFB1 TP53 VEGFA
hsa04657 IL-17 signaling pathway 22 CASP3 CASP8 CHUK MAPK14 FOS GSK3B IFNG IL1B IL4 IL6 CXCL8 JUN MMP1 MMP3 MMP9 NFKBIA MAPK1 PTGS2 RELA CCL2 TNF FOSL1
hsa05145 Toxoplasmosis 22 AKT1 ALOX5 XIAP BCL2 BCL2L1 CASP3 CASP8 CASP9 CD40LG CHUK MAPK14 IFNG IL10 NFKBIA NOS2 PIK3CG MAPK1 RELA STAT1 TGFB1 TNF CYCS
hsa05224 Breast cancer 22 AKT1 BAX CCND1 CDK4 CDKN1A E2F1 E2F2 EGF EGFR ERBB2 ESR1 ESR2 FOS GSK3B JUN MYC PGR MAPK1 PTEN RAF1 RB1 TP53
hsa05170 Human immunodeficiency virus 1 infection 22 AKT1 BAX BCL2 BCL2L1 CASP3 CASP8 CASP9 CCNB1 CDK1 CHEK1 CHUK MAPK14 FOS JUN NFATC1 NFKBIA PRKCA MAPK1 RAF1 RELA TNF CYCS
hsa05022 Pathways of neurodegeneration - multiple diseases 22 BAX BCL2 BCL2L1 CASP3 CASP7 CASP8 CASP9 MAPK14 GSK3B HSPA5 IL1A IL1B IL6 NOS2 PRKCA MAPK1 PTGS2 RAF1 RELA SOD1 TNF CYCS
hsa05219 Bladder cancer 21 CCND1 CDK4 CDKN1A CDKN2A E2F1 E2F2 EGF EGFR ERBB2 CXCL8 MDM2 MMP1 MMP2 MMP9 MYC MAPK1 RAF1 RB1 TP53 VEGFA RASSF1
hsa04115 p53 signaling pathway 21 BAX CCND1 BCL2 BCL2L1 CASP3 CASP8 CASP9 CCNB1 CDK1 CDK2 CDK4 CDKN1A CDKN2A CHEK1 IGFBP3 MDM2 SERPINE1 PTEN TP53 CHEK2 CYCS
hsa04668 TNF signaling pathway 21 AKT1 CASP3 CASP7 CASP8 CHUK MAPK14 FOS ICAM1 IL1B IL6 IRF1 JUN MMP3 MMP9 NFKBIA MAPK1 PTGS2 RELA CCL2 TNF VCAM1
hsa05152 Tuberculosis 21 AKT1 BAX BCL2 CASP3 CASP8 CASP9 MAPK14 CTSD IFNG IL1A IL1B IL6 IL10 NOS2 MAPK1 RAF1 RELA STAT1 TGFB1 TNF CYCS
hsa05132 Salmonella infection 21 AKT1 BAX BCL2 CASP3 CASP7 CASP8 CHUK MAPK14 FOS IL1B IL6 CXCL8 JUN MYC NFKBIA PIK3CG MAPK1 RAF1 RELA TNF CYCS
hsa05142 Chagas disease 20 AKT1 CASP8 CHUK MAPK14 FOS IFNG IL1B IL2 IL6 CXCL8 IL10 JUN NFKBIA NOS2 SERPINE1 MAPK1 RELA CCL2 TGFB1 TNF
hsa04625 C-type lectin receptor signaling pathway 20 AKT1 CASP8 CHUK MAPK14 IL1B IL2 IL6 IL10 IRF1 JUN MDM2 NFATC1 NFKBIA PRKCD MAPK1 PTGS2 RAF1 RELA STAT1 TNF
hsa04510 Focal adhesion 20 AKT1 XIAP CCND1 BCL2 CAV1 EGF EGFR ELK1 ERBB2 FN1 GSK3B JUN KDR MET PRKCA MAPK1 PTEN RAF1 SPP1 VEGFA
hsa05131 Shigellosis 20 AKT1 BAX BCL2 BCL2L1 CHUK MAPK14 EGFR GSK3B HK2 IL1B CXCL8 JUN MDM2 NFKBIA PRKCD MAPK1 RELA TNF TP53 CYCS

In addition, this study constructed a Compound-Target-Pathway network, shown in Figure 7. Analysis of the Compound-Target-Pathway network revealed comprehensive information and intricate relationships that manage cellular activities. Statistical analysis showed that several proteins participated in the top 20 pathways with a high frequency, indicating that they play an important role in the enrichment pathway. The core proteins are TP53, AKT1, VEGFA, JUN, TNF, ESR1, IL-6, and Caspase-3. Therefore, these proteins are critical in the SBD treatment of ovarian cancer.

Figure 7.

Figure 7.

Analysis of Compound-Target-Pathway network. (A) An integrated network of Compound-Target-Pathway of SBD for ovarian cancer: SBD is represented by a hexagon (red), active ingredients by squares (blue), target genes by rhombus (orange), and pathways by arrowheads (green). (B) Network of 20 pathways and their core shared targets: pathways are represented by arrowheads (green) and shared targets by rhombus (orange).

3.2. Molecular docking results and analysis

The interactions between ligands and target proteins is shown in Table 6 and Figure 8. Electrostatic force and van der Waals force are the main forces between ligand and target protein. The lower the binding energy between ligand and receptor was, the more stable the binding conformation and the greater the possibility of interaction. The molecular docking results showed that the binding energy between the effective chemical active ingredient and the key target protein was less than −10 kcal/mol, suggesting that the binding activity between the effective core ingredient and the core target protein is stable.

Table 6.

Docking results of core target proteins with bioactive components.

Core target PDB ID Binding energy/(kcal Mol-1)
Quercetin Luteolin Wogonin
AKT1 1H10 −6.2 −6.0 −5.9
VEGFA 6D3O −6.7 −6.5 −6.5
JUN 1JNM −8.0 −8.1 −6.8
TNF 1A8M −6.6 −7.1 −6.3
Caspase-3 1GFW −6.9 −7.8 −7.3

Figure 8.

Figure 8.

Binding mode of protein and different ligands. (A) Binding mode of AKT1 with quercetin, luteolin and wogonin. (B) Binding mode of Caspase-3 with quercetin, luteolin and wogonin.

3.3. Experimental verification

3.3.1. Quercetin, luteolin, and wogonin inhibits SKOV3 cell proliferation.

The MTT results showed that quercetin, luteolin, and wogonin inhibit the proliferation of SKOV3 cells in a time- and concentration-dependent manner. The study found that the IR of luteolin on the SKOV3 cells was higher than quercetin and wogonin. The results show that the IC50 of quercetin, luteolin, and wogonin in SKOV3 cells treated for 48 hours were 121.43μmol/L, 95.27μmol/L, and 115.57μmol/L respectively, as shown in Figure 9A. Therefore, these concentrations were selected as the intervention concentrations of quercetin, luteolin, and wogonin in subsequent experiments. The plate colony formation assay results show that the colony formation of SKOV3 cells decreased after treatment with quercetin, luteolin, and wogonin than the nontreated groups, as shown in Figure 9B. These results indicate that the 3 core components have an antiproliferative effect on SKOV3 cells. In conclusion, quercetin, luteolin, and wogonin inhibit SKOV3 cell proliferation in a time- and concentration-dependent manner.

Figure 9.

Figure 9.

Quercetin, luteolin, and wogonin inhibit SKOV3 cell proliferation. (A) SKOV3 cells were exposed to a culture medium containing various concentrations of quercetin, luteolin, and wogonin for 24 and 48 hours. The cell viability was measured using the MTT assay. (B) A colony-forming growth assay detection of cell colony formation ability. The colonies were counted and captured. Data are represented as mean ± SD (n = 3). *P < .05, **P < .01, ***P < .001 versus the non-treated group.

3.3.2. Quercetin, luteolin, and wogonin downregulate the mRNA levels of AKT1, VEGFA, JUN, and TNF and upregulate Caspase-3 in SKOV3 cells.

The PPI network screened the top 50 genes, and RT-qPCR verified the expression of 5 key genes. The RT-qPCR results in Figure 10A show that the 5 genes changed as expected after 48 hours of quercetin, luteolin, and wogonin treatment at IC50 concentrations. The mRNA expression levels of AKT1, VEGFA, JUN, and TNF decreased, while the mRNA expression level of Caspase-3 increased in SKOV3 cells treated with quercetin, luteolin, and wogonin than the control group. Moreover, luteolin has the most significant effect on downregulating the expression of AKT1, VEGFA, JUN, and TNF. At the same time, gene expression is shown as a heat map in Figure 10B.

Figure 10.

Figure 10.

(A) RT-qPCR analysis of AKT1, VEGFA, JUN, TNF, and Caspase-3 mRNA expression in SKOV3 cells after stimulation with quercetin(121.43 μmol/L), luteolin (95.27 μmol/L), and wogonin(115.57 μmol/L) for 48 hours. (B) A heat map showing changes in gene expression. Figure 10B is a quantification of Figure 10A. (C, D) Western blotting analysis of markers involved in the PI3K/Akt signaling pathway. Protein levels of PI3K and P-Akt were significantly reduced, and the protein levels of Caspase-3 increased after stimulation with quercetin, luteolin, and wogonin for 48 hours in SKOV3 cells. Data are represented as mean ± SD (n = 3). *P < .05, **P < .01, ***P = .001, ns = no significant differences versus the non-treated group.

3.3.3. Quercetin, luteolin, and wogonin inhibit the activation of the PI3K-Akt signaling pathway.

According to KEGG analysis, the PI3K/Akt signaling pathway was inferred to be the key pathway involved in SBD anti-ovarian cancer pathways. To confirm the KEGG analysis, Western blotting was used to detect the effects of quercetin, luteolin, and wogonin induction of the PI3K/Akt pathway in SKOV3 cells. Figure 10C and 10D show that PI3K and P-Akt were significantly decreased, and the protein levels of Caspase-3 were increased in cells treated with quercetin, luteolin, and wogonin than in the control groups. The results also show that the expression of AKT did not change in the quercetin and luteolin groups. These results suggest that quercetin, luteolin, and wogonin can regulate ovarian cancer by inhibiting the activation of the PI3K/Akt pathway in SKOV3 cells.

4. Discussion

A recent study showed that ovarian cancer (OC) accounted for 3.4% of the 9.2 million new cancer cases in women worldwide in 2020. In addition, OC accounted for 4.7% of 4.4 million female cancer deaths.[31] OC represents the eighth most common cancer and the fifth leading cause of cancer-related deaths among the female population.[32] Although significant progress has been made in treatment of OC, the prognosis remains poor.[33] SBD is a widely used medicinal herb in China because it exerts various medicinal properties, including antioxidative, anti-inflammatory, and anti-cancer effects.[10] The monomer compounds of SBD targeting multiple pathways have been widely studied as antitumor drugs. However, the specific action and mechanism of SBD against ovarian cancer are yet to be clarified.

This study used network pharmacology to explore the molecular mechanism of SBD in treating ovarian cancer. In total, 29 SBD bioactive ingredients were obtained through database screening. The construction of the active ingredients-target interaction network showed that the main active ingredients of SBD were quercetin, luteolin, and wogonin, which are all flavonoids. Studies have shown that flavonoids have significant anticancer effects in various cancer models in vivo and in vitro.[34] Natural flavonoids exert antioxidant, anti-inflammation, and anti-cancerous activities through multiple pathways, and have been shown to induce apoptosis of breast, colorectal, and prostate cancers, lower the nucleoside diphosphate kinase-B activity in lung, bladder, and colon cancers, inhibit cell proliferation and cell cycle arrest in various cancers.[35] Quercetin is the major representative of the flavonoid subclass of flavonols, which widely exists in nuts, teas, vegetables, herbs, and the general daily diet of people.[36] Liu et al investigated the effect of quercetin on apoptosis in mice with ovarian cancer and found that quercetin induced apoptosis via the mitochondria intrinsic and caspase-dependent pathways. Liu et al also reported that quercetin evoked endoplasmic reticulum stress in ovarian cancer resulting in mitochondria-mediated apoptosis.[37] In the xenograft mouse model of ovarian cancer, quercetin treatment increased the anti-tumor effects of cisplatin. In mice treated with cisplatin in combination with pretreatment of quercetin, Bcl-2 expression was downregulated and apoptosis was increased observed compared with other groups. This study suggested that quercetin is a good candidate for adjuvant therapy in ovarian cancer.[38]

On the other hand, related studies have shown that luteolin enhances the anti-tumor effect of cisplatin in ovarian cancer. Combination of luteolin and cisplatin is more effective in suppressing CAOV3/DDP cell growth and metastasis. Therefore, luteolin enhances cisplatin-induced apoptosis in cisplatin-resistant ovarian cancer CAOV3/DDP cells via decreasing Bcl-2 expression.[39] Elsewhere, wogonin treatment increased apoptosis of tumor cells, enhanced the toxicity of TNF-α and TRAIL to tumor cells, blocked the tumor cell cycle, inhibited tumor angiogenesis, and synergized with chemotherapeutic drugs through ROS and Ca2+-mediated signaling pathways.[40] In addition, wogonin acts as an inhibitor of ovary cancer growth. It also induces apoptotic changes and inhibits migration, leading to cell cycle arrest.[41] These active ingredients are the main materials contributing to the anti-ovarian cancer effects of SBD. These ingredients indicate that SBD may produce its anti-ovarian cancer effects through multiple compounds acting on multiple targets and pathways.

In this study, we analyzed active ingredients of SBD and ovarian cancer targets using a PPI network to explore the pharmacological mechanism of SBD in the treatment of ovarian cancer. Consequently, we identified a subset of genes, including TP53, AKT1, VEGFA, JUN, TNF, ESR1, IL-6, and Caspase-3, which exert their effects via multiple different biochemical pathways. Notably, TP53 (p53) is the most widely studied gene in the human genome.[42] Across all human cancers, TP53 is the most commonly altered gene with mutations occurring in approximately 50% of all cancer cases.[43,44] Generally, TP53-mutated tumors exhibit an aggressive phenotype and are characterized by poor differentiation, increased invasiveness, and high metastatic potential. Mutations in TP53 are infrequent in low-grade serous carcinomas or serous borderline tumors but ubiquitous in high-grade serous ovarian cancer, i.e., TP53 mutations in up to 100% of the cases.[45] AKT1 belongs to the protein kinase superfamily and regulates various cellular processes, including cellular metabolism, apoptosis, proliferation, transcription, and migration.[46] AKT1 overexpression or amplification is responsible for the acquired resistance of ovarian cancer cells to paclitaxel.[47] VEGFA is overexpressed in most solid tumors and malignant diseases and significantly stimulates tumor angiogenesis.[48] In addition to affecting endothelial and tumor cells, VEGFA affects tumor function by targeting other cell types in the tumor microenvironment.[49] Previous studies suggest that c-JUN activation subsequently induces both transcription of pro-apoptotic and pro-survival factors. In the context of ovarian cancer, c-JUN enhances the promoter activity of PD-L1 and promotes PD-L1 transcription and expression, which partly explains the upregulation of PD-L1 expression in ovarian cancer.[50] TNF is widely studied in various cancers, including ovarian cancer, and is a major mediator of inflammation. Studies have shown that TNF-α expression is detected by RNA ISH technique in approximately 50% of high-grade serous and 83% of endometrioid ovarian carcinomas.[51] IL-6 is a pleiotropic cytokine and an important biomarker in the cytokine cascade involved in the initiation and regulation of inflammation.[52] IL-6 in ascites and the serum of patients with advanced ovarian cancer has been most strongly correlated with poor survival.[53] ESR1 mutations are associated with resistance to endocrine therapy in metastatic breast cancer and their detection has elicited the development and current evaluation of novel, highly promising therapeutic strategies. In ovarian cancer, ESR1 mutations were detected in 15% of primary tumor tissues and in 13.8% of plasma-cfDNA samples tested using drop-off ddPCR.[54] Caspases are a 15-member family of cysteine proteases, that modulate programmed cell death and inflammation.[55] Caspase-3 activation is critical in breaking down the vital cellular proteins, nuclear fragmentation, and the ultimate death of the cell.[56]

GO functional enrichment and KEGG pathway enrichment analyses found that the cancer pathway, PI3K-Akt signaling pathway and mitogen-activated protein kinase (MAPK) signaling pathway were the most significantly enriched gene sets. Among these, the PI3K/AKT/ pathway is the most frequently altered signaling, found in ~70% of ovarian cancer cases.[57,58] PI3K pathway is frequently upregulated in ovarian cancer and activated PI3K signaling increases cell survival and chemoresistance.[59] MAPK regulates signaling transduction pathways and capacity to control intracellular processes including cell survival, differentiation, proliferation, and apoptosis, via sequential phosphorylation of substrate protein Ser/Thr kinase protein cascades.[60] Activation of the MAPK JNK/p38 pathway is regulated by Gal-1 and facilitates epithelial-mesenchymal transition, suggesting it may be a promising target for the prevention of epithelial ovarian cancer metastasis.[61] Quercetin promotes cell viability loss, apoptosis, and autophagy in cancer by reducing β-catenin and HIF-1α stabilization, inducing caspase-3 activation and inhibiting Akt, mTOR, and ERK phosphorylation. By interfering with PI3K/Akt/mTOR pathways, Quercetin exerts its metabolic effect on cancer, inhibiting key enzymes of glycolysis and glucose uptake.[62] Luteolin inhibits cancer cell proliferation, and survival pathways, including PI3K/Akt, NF-κB, and MAPKs in cancer cells, which may mimic the deprivation of growth factors that blocks the growth factor-triggered signaling pathways.[63] Studies show that wogonin could significantly increase the sensitivity of cisplatin-resistant ovarian cancer cells to cisplatin by downregulating the PI3K/Akt pathway.[64] Therefore, SBD may play an anti-ovarian cancer role via the cancer pathway, the PI3K/Akt signaling pathway, and the MAPK signaling pathway.

This study systematically predicted the material basis and mechanism of SBD in ovarian cancer treatment. In vitro experiments were consistent with the network pharmacology prediction results. Through experiments, the major active ingredients of SBD, i.e., quercetin, luteolin, and wogonin, inhibited proliferation and clone formation, induced apoptosis of SKOV3 cells, regulated mRNA expression of 5 key genes, and inhibited the activity of the PI3K/AKT signaling pathway. This provides a theoretical basis for clinical intervention of ovarian cancer by SBD as well as evidence and references for the development of TCM and monomers for treatment. Therefore, quercetin, luteolin, and wogonin may be ideal drugs for ovarian cancer treatment, however, the underlying molecular mechanism remains to be further studied.

We focused on the study of these 3 different types of small molecular of TCM on ovarian cancer, and the research content in this area is relatively small. In this study, 3 core active components (quercetin, luteolin, wogonin) of SBD were identified. The results of molecular docking has showed a good binding activity between the 3 most important components and the 5 important target proteins (AKT1, VEGFA, JUN, TNF and Caspase-3). And the results of experiments also confirmed that AKT1 and Caspase-3 were the explicit targets and the PI3K/AKT signaling pathway may be the key pathway of the 3 core components in the treatment of ovarian cancer. Therefore, quercetin, luteolin and wogonin are the possible active components of SBD acting on ovarian cancer. However, our study also had certain limitations. Firstly, the current network information technology needs to be further improved, and the accuracy and timeliness of database data require scientific verification. Secondly, the mechanism underlying the effect of SBD on ovarian cancer may be very complicated, the interactions between the multiple components and multiple targets of SBD are also unclear, further in-depth studies will be conducted in the future.

5. Conclusion

Based on the network pharmacology approach, we identified 229 potential active components and 137 related core targets. We screened the key active ingredients, including quercetin, luteolin, wogonin, and hub genes AKT1, VEGFA, JUN, TNF and Caspase-3 of SBD on ovarian cancer. Functional enrichment analysis through KEGG pathways and GO terms revealed that the PI3K/Akt pathway mediated the anti-ovarian cancer effects of SBD. In vitro experiments showed that quercetin, luteolin, and wogonin inhibited the proliferation and induced apoptosis of SKOV3 cells by regulating mRNA expression of 5 key genes, and inhibiting the PI3K/AKT signaling pathway activity.

These results verified the reliability and accuracy of the systematic network pharmacological screening results in this study, suggesting that SBD may potentially treat ovarian cancer, which is worthy of further study. This provides a theoretical basis for the future application of SBD in the treatment of ovarian cancer. Further studies are needed validate our findings.

Author contributions

Data curation: Jie Zhang, Chenhuan Ding.

Project administration: Cong Qi, He Li.

Writing – original draft: Jie Zhang.

Writing – review & editing: Libo Wang, Hongjin Wu, Weiwei Dai, Chenglong Wang.

Supplementary Material

medi-102-e36656-s001.docx (29.9KB, docx)
medi-102-e36656-s002.docx (31.7KB, docx)
medi-102-e36656-s003.docx (28.9KB, docx)
medi-102-e36656-s004.docx (57.2KB, docx)

Abbreviations:

BP
biological processes
CC
cellular components
GO
Gene Ontology
IL-6
interleukin-6
IR
Inhibition Rate
KEGG
Kyoto Encyclopedia of Genes and Genomes
MAPK
mitogen-activated protein kinase
MF
molecular function
OC
ovarian cancer
PPI
protein-protein interaction
SBD
Scutellaria barbata D. Don
TCM
traditional Chinese medicine
TNF
tumor necrosis factor
VEGFA
vascular endothelial growth factor A.

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Supplemental Digital Content is available for this article.

The authors have no conflicts of interest to disclose.

There were no studies involving humans and animals in this study and therefore did not require approval by an ethics committee.

This research was funded by the National Natural Science Foundation of China (82174428 and 81603645).

How to cite this article: Zhang J, Qi C, Li H, Ding C, Wang L, Wu H, Dai W, Wang C. Exploration of the effect and mechanism of Scutellaria barbata D. Don in the treatment of ovarian cancer based on network pharmacology and in vitro experimental verification: Exploration of the effect and mechanism of Scutellaria barbata D. Don in the treatment of ovarian cancer based on network pharmacology and in vitro experimental verification. Medicine 2023;102:51(e36656).

Contributor Information

Cong Qi, Email: qicongxzq@aliyun.com.

He Li, Email: Lihe1972@hotmail.com.

Chenhuan Ding, Email: dingchenhuan@sjtu.edu.cn.

Libo Wang, Email: clwang2018@shutcm.edu.cn.

Hongjin Wu, Email: wuhj130707@shutcm.edu.cn.

Weiwei Dai, Email: wdai2018@shutcm.edu.cn.

Chenglong Wang, Email: clwang2018@shutcm.edu.cn.

References

  • [1].Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin. 2018;68:284–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Chaudhry S, Thomas SN, Simmons GE, Jr. Targeting lipid metabolism in the treatment of ovarian cancer. Oncotarget. 2022;13:768–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Ciucci A, Buttarelli M, Fagotti A, et al. Preclinical models of epithelial ovarian cancer: practical considerations and challenges for a meaningful application. Cell Mol Life Sci. 2022;79:364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Wang S, Long S, Deng Z, et al. Positive role of Chinese herbal medicine in cancer immune regulation. Am J Chin Med. 2020;48:1577–92. [DOI] [PubMed] [Google Scholar]
  • [5].Sheng D, Zhao B, Zhu W, et al. Scutellaria barbata DDon (SBD) extracts suppressed tumor growth, metastasis and angiogenesis in Prostate cancer via PI3K/Akt pathway. BMC Complement Med Ther. 2022;22:120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Wang L, Chen W, Li M, et al. A review of the ethnopharmacology, phytochemistry, pharmacology, and quality control of Scutellaria barbata D Don. J Ethnopharmacol. 2020;254:112260. [DOI] [PubMed] [Google Scholar]
  • [7].Wang Q, Acharya N, Liu Z, et al. Enhanced anticancer effects of Scutellaria barbata D Don in combination with traditional Chinese medicine components on non-small cell lung cancer cells. J Ethnopharmacol. 2018;217:140–51. [DOI] [PubMed] [Google Scholar]
  • [8].Wang Z, Qi F, Cui Y, et al. An update on Chinese herbal medicines as adjuvant treatment of anticancer therapeutics. BioScience Trends. 2018;12:220–39. [DOI] [PubMed] [Google Scholar]
  • [9].Powell CB, Fung P, Jackson J, et al. Aqueous extract of herba Scutellaria barbatae, a chinese herb used for ovarian cancer, induces apoptosis of ovarian cancer cell lines. Gynecol Oncol. 2003;91:332–40. [DOI] [PubMed] [Google Scholar]
  • [10].Zhang L, Ren B, Zhang J, et al. Anti-tumor effect of Scutellaria barbata D Don extracts on ovarian cancer and its phytochemicals characterisation. J Ethnopharmacol. 2017;206:184–92. [DOI] [PubMed] [Google Scholar]
  • [11].Song YJ, Bao JM, Zhou LY, et al. An analysis of the anti-neuropathic effects of qi she pill based on network pharmacology. Evid Based Complement Alternat Med. 2020;2020:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Song Y, Wang H, Pan Y, et al. Investigating the multi-target pharmacological mechanism of Hedyotis diffusa willd acting on prostate cancer: a network pharmacology approach. Biomolecules. 2019;9:591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Ren B, Tan L, Xiong Y, et al. Integrated analysis of the mechanisms of da-chai-hu decoction in type 2 diabetes mellitus by a network pharmacology approach. Evid Based Complement Alternat Med. 2020;2020:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Meng Z, Liu X, Wu J, et al. Mechanisms of compound kushen injection for the treatment of lung cancer based on network pharmacology. EvidBased Complement Alternat Med. 2019;2019:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Ru J, Peng L, Jinan W, et al. TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. J Cheminf. 2014;6:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Kim S, Thiessen PA, Bolton EE, et al. PubChem substance and compound databases. Nucleic Acids Res. 2016;44(D1):D1202–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Gfeller D, Grosdidier A, Wirth M, et al. SwissTargetPrediction: a web server for target prediction of bioactive small molecules. Nucleic Acids Res. 2014;42(W1):W32–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Wang X, Shen Y, Wang S, et al. PharmMapper 2017 update: a web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Res. 2017;45(W1):W356–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].UniProt Consortium. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res. 2021;49(D1):D480–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Wishart DS, Feunang YD, Guo AC, et al. DrugBank 50: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48(D1):D845–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Stelzer G, Rosen N, Plaschkes I, et al. The GeneCards suite: From gene data mining to disease genome sequence analyses. Curr Protoc Bioinform. 2016;54:30–1. [DOI] [PubMed] [Google Scholar]
  • [23].Amberger JS, Bocchini CA, Schiettecatte F, et al. OMIMorg: Online mendelian inheritance in man (OMIM(R)), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43:D789–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Bardou P, Mariette J, Escudié F, et al. jvenn: an interactive Venn diagram viewer. BMC Bioinf. 2014;15:293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Dong C, Tian X, He F, et al. Integrative analysis of key candidate genes and signaling pathways in ovarian cancer by bioinformatics. J Ovarian Res. 2021;14:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Zhou Y, Zhou B, Pache L, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Goodsell DS, Zardecki C, di Costanzo L, et al. RCSB protein data bank: enabling biomedical research and drug discovery. Protein science. 2020;29:52–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].El-Hachem N, Haibe-Kains B, Khalil A, et al. AutoDock and AutoDockTools for protein-ligand docking: beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) as a case study. Methods Mol Biol. 2017;1598, article 28508374:391–403. [DOI] [PubMed] [Google Scholar]
  • [30].Baugh EH, Lyskov S, Weitzner BD, et al. Real-time PyMOL visualization for Rosetta and PyRosetta. PLoS One. 2011;6:e21931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49. [DOI] [PubMed] [Google Scholar]
  • [32].Cao W, Chen HD, Yu YW, et al. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J (Engl). 2021;134:783–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Rodel F, Zhou S, Gyorffy B, et al. The prognostic relevance of the proliferation markers Ki-67 and Plk1 in early-stage ovarian cancer patients with serous, low-grade carcinoma based on mRNA and protein expression. Front Oncol. 2020;10:558932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Wen K, Fang X, Yang J, et al. Recent research on flavonoids and their biomedical applications. Curr Med Chem. 2021;28:1042–66. [DOI] [PubMed] [Google Scholar]
  • [35].Hazafa A, Rehman KU, Jahan N, et al. The role of polyphenol (Flavonoids) compounds in the treatment of cancer cells. Nutr Cancer. 2020;72:386–97. [DOI] [PubMed] [Google Scholar]
  • [36].Metodiewa D, Jaiswal AK, Cenas N, et al. Quercetin may act as a cytotoxic prooxidant after its metabolic activation to semiquinone and quinoidal product. Free Radic Biol Med. 1999;26:107–16. [DOI] [PubMed] [Google Scholar]
  • [37].Liu Y, Gong W, Yang ZY, et al. Quercetin induces protective autophagy and apoptosis through ER stress via the p-STAT3/Bcl-2 axis in ovarian cancer. Apoptosis. 2017;22:544–57. [DOI] [PubMed] [Google Scholar]
  • [38].Yang Z, Liu Y, Liao J, et al. Quercetin induces endoplasmic reticulum stress to enhance cDDP cytotoxicity in ovarian cancer: involvement of STAT3 signaling. FEBS J. 2015;282:1111–25. [DOI] [PubMed] [Google Scholar]
  • [39].Wang H, Luo Y, Qiao T, et al. Luteolin sensitizes the antitumor effect of cisplatin in drug-resistant ovarian cancer via induction of apoptosis and inhibition of cell migration and invasion. J Ovarian Res. 2018;11:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Xiao W, Bu P, Gong W. Research progress on anti-tumor and immunomodulatory effects of wogonin. China J Chin Mater Med. 2014;39:3004–9. [PubMed] [Google Scholar]
  • [41].Zhao Q, Chang W, Chen R, et al. Anti-Proliferative effect of wogonin on ovary cancer cells involves activation of apoptosis and cell cycle arrest. Med Sci Monit. 2019;25:8465–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Dolgin E. The most popular genes in the human genome. Nature. 2017;551:427–31. [DOI] [PubMed] [Google Scholar]
  • [43].Levine AJ. Spontaneous and inherited TP53 genetic alterations. Oncogene. 2021;40:5975–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Priestley P, Baber J, Lolkema MP, et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature. 2019;575:210–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Silwal-Pandit L, Langerød A, Børresen-Dale AL. TP53 mutations in breast and ovarian cancer. Cold Spring Harb Perspect Med. 2017;7:a026252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017;169:381–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Page C, Lin HJ, Jin Y, et al. Overexpression of Akt/AKT can modulate chemotherapy-induced apoptosis. Anticancer Res. 2000;20:407–16. [PubMed] [Google Scholar]
  • [48].Claesson-Welsh L, Welsh M. VEGFA and tumour angiogenesis. J Intern Med. 2013;273:114–27. [DOI] [PubMed] [Google Scholar]
  • [49].Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013;13:871–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Yang Y, Xia L, Wu Y, et al. Programmed death ligand-1 regulates angiogenesis and metastasis by participating in the c-JUN/VEGFR2 signaling axis in ovarian cancer. Cancer Commun (Lond). 2021;41:511–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Gupta M, Babic A, Beck AH, et al. TNF-α expression, risk factors, and inflammatory exposures in ovarian cancer: evidence for an inflammatory pathway of ovarian carcinogenesis? Hum Pathol. 2016;54:82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Kishimoto T. Interleukin-6: discovery of a pleiotropic cytokine. Arthritis Res Ther. 2006;8(Suppl 2(Suppl 2):S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Kampan NC, Madondo MT, McNally OM, et al. Interleukin 6 present in inflammatory ascites from advanced epithelial ovarian cancer patients promotes tumor necrosis factor receptor 2-expressing regulatory T cells. Front Immunol. 2017;8:1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Stergiopoulou D, Markou A, Giannopoulou L, et al. Detection of ESR1 mutations in primary tumors and plasma cell-free DNA in high-grade serous ovarian carcinoma patients. Cancers (Basel). 2022;14:3790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Eckhart L, Ballaun C, Uthman A, et al. Identification and characterization of a novel mammalian caspase with proapoptotic activity. J Biol Chem. 2005;280:35077–80. [DOI] [PubMed] [Google Scholar]
  • [56].Nagraj J, Chatterjee S, Pal T, et al. A novel series of di-fluorinated propanedione derivatives synergistically augment paclitaxel mediated caspase 3 activation in ovarian cancer cells. J Cancer Res Ther. 2014;10:701–9. [DOI] [PubMed] [Google Scholar]
  • [57].Lheureux S, Gourley C, Vergote I, et al. Epithelial ovarian cancer. Lancet. 2019;393:1240–53. [DOI] [PubMed] [Google Scholar]
  • [58].Ediriweera MK, Tennekoon KH, Samarakoon SR. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin Cancer Biol. 2019;59:147–60. [DOI] [PubMed] [Google Scholar]
  • [59].Huang TT, Lampert EJ, Coots C, et al. Targeting the PI3K pathway and DNA damage response as a therapeutic strategy in ovarian cancer. Cancer Treat Rev. 2020;86:102021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Katopodis P, Kerslake R, Zikopoulos A, et al. p38β - MAPK11 and its role in female cancers. J Ovarian Res. 2021;14:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Zhu J, Zheng Y, Zhang H, et al. Galectin-1 induces metastasis and epithelial-mesenchymal transition (EMT) in human ovarian cancer cells via activation of the MAPK JNK/p38 signalling pathway. Am J Transl Res. 2019;11:3862–78. [PMC free article] [PubMed] [Google Scholar]
  • [62].Reyes-Farias M, Carrasco-Pozo C. The anti-cancer effect of quercetin: molecular implications in cancer metabolism. Int J Mol Sci . 2019;20:3177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Lin Y, Shi R, Wang X, et al. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr Cancer Drug Targets. 2008;8:634–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Xing F, Sun C, Luo N, et al. Wogonin increases cisplatin sensitivity in ovarian cancer cells through inhibition of the phosphatidylinositol 3-Kinase (PI3K)/Akt pathway. Med Sci Monit. 2019;25:6007–14. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

medi-102-e36656-s001.docx (29.9KB, docx)
medi-102-e36656-s002.docx (31.7KB, docx)
medi-102-e36656-s003.docx (28.9KB, docx)
medi-102-e36656-s004.docx (57.2KB, docx)

Articles from Medicine are provided here courtesy of Wolters Kluwer Health

RESOURCES