Play all audios:
ABSTRACT Sanbi Decoction (SBD) has demonstrated promising therapeutic potential in osteoarthritis (OA) treatment, yet its precise mechanisms remain unclear. This research combined
computational and experimental approaches, including bioinformatics analysis, network pharmacology, molecular docking, molecular dynamics simulations, and laboratory validation, to
investigate the mechanisms of action of SBD. A total of 114 active compounds and 113 intersecting targets were identified through TCMSP and multiple screening strategies. Gene Ontology (GO)
and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that these targets are primarily involved in key signaling pathways, including the AGE-RAGE signaling pathway,
IL-17 signaling pathway, and TNF signaling pathway. Among the active components, Shinflavanone, Gancaonin L, Xambioona, Phaseol, Gancaonin O, and Licoisoflavanone exhibited strong binding
affinity and structural stability with core targets, as validated by molecular docking and molecular dynamics simulations. Experimental results confirmed that SBD alleviates oxidative
stress, reduces inflammation, and protects cartilage by inhibiting the AGE-RAGE/JNK pathway. These findings highlight SBD’s potential as a promising therapeutic agent for OA treatment.
SIMILAR CONTENT BEING VIEWED BY OTHERS BERGENIN PROTECTS AGAINST OSTEOARTHRITIS BY INHIBITING STAT3, NF-ΚB AND JUN PATHWAYS AND SUPPRESSING OSTEOCLASTOGENESIS Article Open access 31 August
2024 EXPLORING THE ANTI-INFLAMMATORY ACTIVITY OF FUPENZIC ACID USING NETWORK PHARMACOLOGY AND EXPERIMENTAL VALIDATION Article Open access 24 April 2025 PROBING THE MOLECULAR MECHANISM OF
KAEMPFEROL IN RELIEVING RHEUMATOID ARTHRITIS BASED ON NETWORK PHARMACOLOGY Article Open access 12 April 2025 INTRODUCTION Osteoarthritis (OA) is a prevalent degenerative joint disease
characterized by dysfunction, pain, and stiffness1. Synovitis is a key pathological feature of OA, which severely impacts patients’ quality of life2. According to the Global Burden of
Disease study, OA is a major public health concern, with 607 million people affected worldwide in 20213. The dual physical and psychological burden of OA imposes significant challenges on
families and society. Traditional Chinese Medicine (TCM) has demonstrated unique advantages in the treatment of OA due to its efficacy and minimal side effects, contributing significantly to
global health4,5. TCM approaches to treating OA are multifaceted, focusing on promoting blood circulation, nourishing the liver and kidneys, and improving joint function6,7. Sanbi Decoction
(SBD) is a traditional herbal formulation created by Dr. Li Baochao, a distinguished practitioner of Traditional Chinese Medicine (TCM) from Luoyang, China. This formula consists of eight
herbs: Radix Saposhnikoviae (Fangfeng), Notopterygium Root (Qianghuo), Radix Gentianae Macrophyllae (Qinjiao), Coix Seed (Yiyiren), Angelica Sinensis (Danggui), Radix Aconiti (Chuanwu),
Radix Aconiti Kusnezoffii (Caowu), and Glycyrrhizae Radix (Gancao). SBD is known for its effects in dispelling wind and dampness and strengthening tendons and bones. While its components or
their compounds have shown potential for treating OA, the specific mechanisms and targets remain unclear, necessitating more comprehensive insights. Embracing novel approaches and fresh
insights is essential for gaining a more comprehensive understanding of the anti-osteoarthritis mechanisms of Traditional Chinese Medicine formulations. In recent years, with the rapid
development of bioinformatics, network pharmacology combined with computational simulations has emerged as a promising approach, laying the foundation for comprehensively understanding the
mechanisms of TCM in disease treatment8,9,10. As a computational technique, molecular docking predicts the binding affinity and interaction patterns between small-molecule ligands from
Traditional Chinese Medicine and protein receptors. This approach offers valuable insights into drug-target interactions and supports the pharmacological assessment of both TCM formulations
and individual herbs11. However, molecular docking lacks detailed assessments of interaction stability and dynamics. Molecular dynamics (MD) simulations play a critical role in
bioinformatics, particularly in analyzing ligand-receptor interactions12. These simulations provide in-depth insights into the dynamic behavior of biomolecules, uncovering key information
about binding mechanisms, stability, and conformational changes. For instance, MD studies on G protein-coupled receptors (GPCRs) have revealed important insights into their conformational
dynamics, essential for understanding their role as drug targets13. Similarly, MD simulations have been pivotal in uncovering hidden binding sites and predicting drug-binding poses when
targeting small GTPases, crucial for developing novel inhibitors14. The integration of network pharmacology, molecular docking, MD simulations, and experimental validation enhances the
reliability of results15,16. Therefore, employing these methodologies to identify the active components and targets of SBD holds great potential for elucidating its anti-OA mechanisms (Fig.
1). MATERIALS AND METHODS ACQUISITION AND ORGANIZATION OF GEO DATA Gene expression profile datasets GSE55235 and GSE55457 were retrieved from the GEO (Gene Expression Omnibus,
https://www.ncbi.nlm.nih.gov/geo/) database using “osteoarthritis” as the keyword. These datasets encompass microarray data from 50 samples. DATA ANALYSIS USING PERL AND R Background
correction and matrix normalization were performed in the R environment (version 4.4.1: https://www.r-project.org/) based on the characteristics of the data samples. The Limma R package
(version 3.62.1, https://bioconductor.org/packages/limma/) was utilized to analyze differential gene expression in the microarray data. Significant differentially expressed genes (DEGs) were
identified using filtering criteria of an adjusted p-value < 0.05 and an absolute value of log fold change (| log FC |) ≥ 1. Volcano plots for the microarray data were generated using
the ggplot2 package (version 3.5.1, https://ggplot2.tidyverse.org/), and heatmaps of the gene chips were created using the pheatmap package (version 1.0.12,
https://cran.r-project.org/package=pheatmap). SELECTION OF SBD ACTIVE COMPONENTS AND TARGETS The herbal composition of SBD was input into the TCMSP (Traditional Chinese Medicine Systems
Pharmacology Database and Analysis Platform, https://www.91tcmsp.com/#/database). Active components were filtered using thresholds of oral bioavailability (OB) ≥ 30% and drug-likeness (DL) ≥
0.1817,18, along with their corresponding targets. The UniProt (Universal Protein Resource, https://www.uniprot.org/) was used for target validation by confirming gene names and UniProt
IDs. Non-human genes were excluded to ensure the precise identification of potential targets of SBD active components. IDENTIFICATION OF POTENTIAL DISEASE TARGETS For osteoarthritis-related
research, target information was collected from GeneCards (GeneCards® Human Gene Database, https://www.genecards.org/), DisGeNET (Disease-Gene Association Network,
https://www.disgenet.org/), the Online Mendelian Inheritance in Man database (OMIM: https://www.omim.org/), and the Therapeutic Target Database (TTD: https://db.idrblab.net/ttd/). These
targets were merged and deduplicated using the UniProt database. The results were then combined with differentially expressed genes (DEGs) from the GEO database, with duplicate entries
removed to establish a comprehensive disease target dataset. CONSTRUCTION OF PROTEIN-PROTEIN INTERACTION NETWORK AND CORE TARGET SCREENING The intersection of potential targets for SBD and
OA was identified using the Venn platform, generating a Venn diagram. A protein-protein interaction (PPI) network for intersecting genes was constructed using the STRING (Search Tool for the
Retrieval of Interacting Genes/Proteins, https://cn.string-db.org/), with “Homo sapiens” as the target species and a confidence threshold of “high confidence” (≥ 0.700)19,20. Finally,
Cytoscape (version 3.10.2, https://cytoscape.org/) and the CytoHubba plugin were employed to analyze three key metrics: degree centrality (Degree), maximal clique centrality (MCC), and
network centrality (MNC). The six highest-ranked genes were selected to establish the core target network. CONSTRUCTION OF THE DRUG-ACTIVE COMPONENT-COMMON TARGET NETWORK A network diagram
was generated using Cytoscape to depict the interactions among drugs, active components, and shared targets. This visualization enhances the understanding of compound-target relationships
and their potential mechanisms of action. GO AND KEGG ENRICHMENT ANALYSIS GO (Gene Ontology, http://geneontology.org/) and KEGG (Kyoto Encyclopedia of Genes and Genomes,
https://www.genome.jp/kegg/) enrichment analyses were conducted in the R environment. The org.Hs.eg.db package (version 3.2.0, https://bioconductor.org/packages/org.Hs.eg.db/) was employed
for gene annotation, while the clusterProfiler package (version 4.14.4, https://bioconductor.org/packages/clusterProfiler/) facilitated gene ID conversion and pathway enrichment analysis of
the identified common targets21. These methods accurately identified GO terms related to biological processes, molecular functions, and cellular components associated with SBD. KEGG pathway
enrichment analysis was also performed22. The results were visualized using the ggplot2 package, offering a clear presentation of enriched GO terms and KEGG pathways to support further
research. TARGET-KEGG NETWORK CONSTRUCTION A network illustrating interactions between common targets and KEGG pathways was constructed using Cytoscape. The “Network Analyzer” module was
used to analyze network properties. Targets meeting three critical indicators—Degree, MCC, and MNC—were identified as core targets. MOLECULAR DOCKING SIMULATION The full names of key target
proteins were verified using the UniProt database, and their corresponding three-dimensional (3D) structures were obtained from the PDB (Protein Data Bank, https://www.rcsb.org/) database.
The selected protein structures included IL1B (PDB ID: 8RYS), IL6 (PDB ID: 1IL6), TNF (PDB ID: 1TNF), JNK (PDB ID: 4QTD), and STAT3 (PDB ID: 6NJS). Using PyMOL (version 3.1,
https://pymol.org/), water molecules were removed from the protein structures. The three-dimensional (3D) molecular structures of key active compounds were retrieved from the PubChem and
TCMSP databases. Using OpenBabel (version 2.3.2, https://openbabel.org/), these structures were converted from SDF format to mol2 format for molecular docking analysis. Hydrogen atoms were
added, and charges were calculated with AutoDockTools (version 1.5.7, http://autodock.scripps.edu/resources/adt) to prepare the proteins for molecular docking23. After target annotation,
docking was conducted using AutoDockTools24. Binding energy heatmaps were generated using the ggplot2 package in R to illustrate the docking affinities. To validate the RMSD values, ensure
that the RMSD is less than 2.0 Å to confirm the reliability of the docking results. Two-dimensional interaction diagrams were generated using Discovery Studio Visualizer (version 19.1,
https://discover.3ds.com/discovery-studio-visualizer-download)25,26. Finally, 3D images were generated using PyMOL. MOLECULAR DYNAMICS SIMULATION Based on the molecular docking results from
AutoDock, the top-ranked model was selected as the starting point for molecular dynamics (MD) simulations. The Amber99sb force field was used to characterize proteins, while the GAFF force
field was applied to ligands. Topology files for each ligand were generated using the Sobtop tool (v.3.1, https://www.sobtop.com/). MD simulations were conducted using GROMACS (version
2020.6, http://www.gromacs.org/)27, with each complex solvated in an SPC water model and neutralized with sodium and chloride ions. Energy minimization was conducted to optimize the system,
followed by a gradual temperature increase to 300 K over 200 ns. Subsequently, a 2-ns NPT equilibration was performed at 1 bar to stabilize the system. Finally, a 200-ns production molecular
dynamics simulation was executed under controlled conditions of 300 K and 1 bar, with a timestep of 2 fs. The cutoff distance for van der Waals and short-range electrostatic interactions
was set at 1.0 nm. The LINCS algorithm was used to constrain hydrogen bonds, with V-rescale and Berendsen methods employed to maintain stable temperature and pressure conditions. During the
MD simulation, several parameters were analyzed to assess the conformational stability of the protein-ligand complex, including root mean square deviation (RMSD), root mean square
fluctuation (RMSF), solvent-accessible surface area (SASA), radius of gyration (Rg), hydrogen bonds (HB), and principal component analysis (PCA). The MM/PBSA (Molecular
Mechanics/Poisson-Boltzmann Surface Area) method was applied to calculate the binding free energy, providing insights into the thermodynamic stability of the complex. DRUG PREPARATION AND
CELL CULTURE The eight herbal components of SBD were ground into a fine powder in proportion and dissolved in distilled water at a concentration of 1 g/mL, The mixture was then centrifuged
at 5000 rpm for 20 min. The supernatant was collected and filtered using a 0.22 μm microporous membrane to obtain the 1 g/mL crude extract, which was stored at − 80 °C. Murine
macrophage-like RAW264.7 cells (Catalog No. JW-CL-0582) were purchased from Shanghai Jiwei Biotechnology Co., Ltd. The cells were cultured in DMEM high-glucose medium supplemented with 10%
fetal bovine serum (FBS) under standard conditions of 37 °C with 5% CO₂. CCK-8 CELL VIABILITY ASSAY Cell viability was assessed using the Cell Counting Kit-8 (CCK-8, Catalog No. CK04-500).
Briefly, RAW264.7 cells were seeded into 96-well plates at a density of 7.5 × 10³ cells/well and incubated for 24 h. The cells were then treated with SBD at concentrations of 12.5, 25, 50,
100, 200, and 400 µg/mL for 24 h. After treatment, 10µL of CCK-8 reagent was added to each well, followed by incubation at 37 °C for 1 h. The optical density (OD) was measured at 450 nm
using a microplate reader. Cell viability was calculated using the standard formula, and the experimental data were visualized using GraphPad Prism 9.5. REAL-TIME QUANTITATIVE PCR (RT-QPCR)
ANALYSIS OF GENE EXPRESSION RAW264.7 cells were seeded into 6-well plates and incubated for 24 h. Cells in the control group were treated with phosphate-buffered saline (PBS), while the OA
group and SBD group were stimulated with LPS (100 ng/mL) for 2 h. Subsequently, the SBD group was treated with SBD for 24 h. After treatment, the culture medium was discarded, and the cells
were washed with PBS. Total RNA was extracted using an RNA extraction kit, followed by reverse transcription into cDNA using a reverse transcription kit. qPCR was performed using cDNA as a
template on the LightCycler®96 system with the following primer sequences: IL1-β: Forward: 5′-TGCCACCTTTTGACAGTGATG-3′. Reverse: 5′-ATGTGCTGCTGCGAGATTTG-3′. IL-6: Forward:
5′-GGAGCCCACCAAGAACGATAG-3′. Reverse: 5′-GTGAAGTAGGGAAGGCCGTG-3′. TNF: Forward: 5′-CCCTCACACTCAGATCATCTTCT-3′. Reverse: 5′-GCTACGACGTGGGCTACAG-3′. STAT3: Forward: 5′-CCGCAGCTTGGGCTGGAAGA-3′.
Reverse: 5′-CAGGGCCGGGCTGTGGTAGT-3′. JNK: Forward: 5′-GTGGAATCAAGCACCTTCACT-3′. Reverse: 5′-TCCTCGCCAGTCCAAAATCAA-3′. GAPDH (Internal control): Forward: 5′-TGGATTTGGACGCATTGGTC-3′. Reverse:
5′-TTTGCACTGGTACGTGTTGAT-3′. All primers were synthesized by Nanning GenSys Biotechnology Co., Ltd. The 2−∆∆Ct method was used to calculate the fold change in gene expression, normalized to
GAPDH. Experimental data were visualized using GraphPad Prism 9.5 software. DETECTION OF ROS SCAVENGING ABILITY USING THE DCFH PROBE RAW264.7 cells were seeded into 12-well plates and
incubated for 24 h. The grouping and treatment methods were the same as in Section “Real-time quantitative PCR (RT-qPCR) analysis of gene expression”. After treatment, the cells were washed
with phosphate-buffered saline (PBS) and incubated with the DCFH-DA fluorescent probe. Fluorescence imaging was performed using a FITC channel, and fluorescence intensity was quantified
using ImageJ software, normalized, and visualized using GraphPad Prism 9.5 software. IMMUNOFLUORESCENCE STAINING RAW264.7 cells were seeded into 12-well plates and incubated for 24 h. The
grouping and treatment methods were the same as in Section “Real-time quantitative PCR (RT-qPCR) analysis of gene expression”. After treatment, the cells were washed with PBS, fixed with 4%
paraformaldehyde, permeabilized with Triton X-100, and blocked with a blocking solution. Cells were then incubated with JNK Polyclonal antibody (Cat No. 51153-1-AP), followed by incubation
with a FITC-labeled secondary antibody (BA1105) for fluorescence detection. Finally, nuclei were counterstained with DAPI. Fluorescence images were captured under a fluorescence microscope,
and fluorescence intensity was quantified using ImageJ software, normalized, and visualized using GraphPad Prism 9.5 software. WESTERN BLOT ANALYSIS RAW264.7 cells were seeded into 6-well
plates and incubated for 24 h under standard conditions. Cell grouping and treatment were performed as described in Section “Real-time quantitative PCR (RT-qPCR) analysis of gene
expression”. Following treatment, cells were washed with PBS, and total protein was extracted using RIPA lysis buffer supplemented with PMSF and phosphatase inhibitors. The protein lysates
were separated via SDS-PAGE, transferred onto a PVDF membrane, and subsequently blocked. The membrane was then incubated with a JNK polyclonal antibody (Cat No. 51153-1-AP), followed by an
HRP-conjugated goat anti-rabbit secondary antibody (Cat No. SA00001-2) after PBS washing. Protein bands were detected using an enhanced chemiluminescence (ECL) system, and their intensities
were quantified with ImageJ software. The data were then normalized and graphically presented using GraphPad Prism 9.5. RESULTS IDENTIFICATION OF DIFFERENTIALLY EXPRESSED GENES (DEGS) After
data preprocessing, 432 differentially expressed genes (DEGs) were identified in osteoarthritis (OA) tissues (Fig. 2A). Among them, 199 genes were downregulated, and 233 genes were
upregulated. The top 50 most significantly upregulated and downregulated genes were selected for further analysis (Fig. 2B). IDENTIFICATION OF ACTIVE COMPONENTS AND THEIR CORRESPONDING
TARGETS IN SBD By integrating data from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and eliminating duplicates, 114 potentially active compounds were identified
across different herbal sources: Aconite (2 compounds), Angelica sinensis (2 compounds), Saposhnikovia divaricata (18 compounds), Coix lacryma-jobi (6 compounds), Glycyrrhiza uralensis (87
compounds), Notopterygium incisum (13 compounds), Gelsemium elegans (1 compound), and Gentiana macrophylla (1 compound). Utilizing the UniProt database for subsequent verification and
refinement, we ultimately identified targets from Aconite (4 targets), Angelica sinensis (42 targets), Saposhnikovia divaricata (64 targets), Coix lacryma-jobi (27 targets), Glycyrrhiza
uralensis (199 targets), Notopterygium incisum (37 targets), Gelsemium elegans (19 targets), and Gentiana macrophylla (27 targets). For additional details, please consult the supplementary
materials. POTENTIAL OA-RELATED TARGET GENES Using databases such as GeneCards, DISGENET, OMIM, and TTD, 2,046 target genes related to OA were identified after removing duplicates (Fig. 3A).
CONSTRUCTION OF PPI NETWORK Venn diagram analysis identified 113 overlapping targets between SBD and OA, highlighting potential therapeutic targets (Fig. 3B). These targets were imported
into the STRING database with a high-confidence threshold of 0.700 for PPI network construction (Fig. 3C). Analysis using Cytoscape (v3.10.2) revealed 107 genes and 930 edges in the network.
The CytoHubba plugin identified key genes based on Degree, MCC, and MNC metrics (Figs. 3D–F). DRUG-ACTIVE INGREDIENT-COMMON TARGET NETWORK To clarify the interactions between SBD active
compounds and their targets, a drug-active ingredient-target network was established (Fig. 4). This network comprised 237 nodes, including 1 formula name, 1 disease name, 8 herbal sources,
114 active compounds, and 113 target genes, interconnected by 948 edges. GO AND KEGG PATHWAY ENRICHMENT ANALYSIS To gain a clear understanding of the functions of the intersecting genes, GO
(Gene Ontology) functional enrichment and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analyses were conducted on 113 intersecting targets using R software (v4.4.1) with
a significance threshold of _P_ < 0.05. GO analysis revealed that the biological processes (BP) were predominantly associated with antioxidant responses, TNF-mediated signaling pathways,
cellular responses to bacterial components and lipopolysaccharides, as well as inflammation-related mechanisms, including cell proliferation and the regulation of apoptotic signaling
pathways. The cellular component (CC) was mainly regions such as membrane rafts, membrane microdomains, vesicular lumens, secretory granule lumens, and the outer mitochondrial membrane; the
molecular function (MF) was primarily related to ligand activity, DNA-binding transcription factor binding, protein kinase regulatory activity, and receptor binding functions. The top 10
terms for biological processes (BP), cellular components (CC), and molecular functions (MF) were visualized using multiple chart formats with the ggplot2 package (Fig. 5A). KEGG pathway
analysis identified 175 significantly enriched pathways (_P_ < 0.05), with key pathways including lipid metabolism and atherosclerosis, the RAGE signaling pathway in AGE-related diabetic
complications, the IL-17 signaling pathway, and the TNF signaling pathway. The top 30 pathways were visualized using the ggplot2 package (Fig. 5B). The roles of the AGE-RAGE signaling
pathway and IL-17 signaling pathway are particularly significant in the regulation of inflammation and oxidative stress by SBD in OA (Fig. 5C,D). TARGET-KEGG NETWORK CONSTRUCTION To clarify
the interconnections between SBD targets and pathways in OA treatment, we established a drug-active compound-common target-KEGG network. This network consisted of 267 nodes, including 4 core
targets, 109 additional targets, 30 pathways, and 1,608 edges (Fig. 6). Among them, the 4 core targets, which hold significant positions in Degree, MCC, and MNC analyses, were selected for
subsequent molecular docking studies. MOLECULAR DOCKING The core active compounds (Shinflavanone, Gancaonin L, Xambioona, Phaseol, Gancaonin O, and Licoisoflavanone) were used as ligands,
while key therapeutic proteins (IL6, TNF, STAT3, IL1B, and JNK) were chosen based on their hydrogen bonding interactions and binding energies. The results were displayed as binding energy
heatmaps using ggplot2 (Fig. 7A). Notably, JNK acts as an upstream regulator of IL6, TNF, and IL1B. Shinflavanone showed the lowest binding energy with IL1B, JNK, and STAT3, mainly
interacting through hydrogen bonds (VAL A:41; ASN A:114) and Van der Waals forces (CYS A:251; ASP A:334) in the binding pocket. Xambioona had the strongest affinity for IL6, stabilizing the
protein-ligand complex via hydrogen bonding (VAL A:97) and Van der Waals interactions (LEU A:93; ASN A:145). Gancaonin L exhibited the lowest binding energy with TNF, forming a stable
complex primarily through Van der Waals forces (SER B:99) (see Supplementary Material S4 for details). The best-performing candidates for each complex were visualized using PyMOL and
Discovery Studio (Fig. 7B–E). Additionally, three independent docking repetitions produced consistent and stable conformations, validating the reliability of molecular docking performed with
AutoDockTools. MD SIMULATION Based on the molecular docking results, 200-ns MD simulations were performed for the following protein-ligand complexes: Xambioona_IL6, Gancaonin L_TNF,
Shinflavanone_STAT3, Shinflavanone_JNK, and Shinflavanone_IL1B. The stability of the trajectory files was assessed using multiple metrics, including root-mean-square deviation (RMSD),
root-mean-square fluctuation (RMSF), radius of gyration (Rg), solvent-accessible surface area (SASA), hydrogen bond dynamics (HB), and principal component analysis-free energy landscape
(PCA). As shown in Fig. 8A, the RMSD variations for all ligands remained below 0.1 nm, indicating high stability of the ligands throughout the simulation. The RMSD profile of the
Shinflavanone_IL1B complex exhibited noticeable fluctuations, suggesting structural and conformational changes due to protein folding. In contrast, the other complexes displayed relatively
stable RMSD profiles throughout the simulation. As shown in Fig. 8B–D, the Shinflavanone_IL1B complex demonstrated increased flexibility in two regions (31–38 and 48–56), while the
Shinflavanone_STAT3 complex exhibited similar flexibility in the 170–220 region. The Xambioona_IL6 complex displayed flexibility in three regions (42–70, 71–81, and 130–170). The Rg and SASA
analyses revealed low fluctuation levels, indicating greater overall structural stability of the complexes. HB analysis revealed fewer than four hydrogen bonds between the molecules,
suggesting that the interactions were primarily driven by van der Waals forces, which was further corroborated by the subsequent MM-PBSA analysis (Fig. 8E). PCA plots during molecular
dynamics simulation revealed the dominant conformational changes and dynamic behaviors of the protein throughout the simulation (Fig. 8F). The MM-PBSA results, used to quantify the binding
free energy of the complexes (see Supplementary Material S5 for details), confirmed that all ligand-protein complexes exhibited favorable binding affinity. EFFECTS OF SBD ON CELL VIABILITY
As shown in Fig. 9A, CCK-8 assay results indicate that SBD treatment does not exhibit significant cytotoxicity at concentrations up to 100 µg/mL, However, at 200 µg/mL, a noticeable decrease
in cell viability was observed. In the concentration range of 12.5–100 µg/mL, cell viability remained stable with a slight proliferative effect. Therefore, 100 µg/mL was selected as the
non-cytotoxic concentration for subsequent experiments, ensuring both safety and therapeutic efficacy. EFFECTS OF SBD ON INFLAMMATORY MEDIATORS AND ROS QRT-PCR analysis revealed that LPS
stimulation significantly induced the expression of inflammatory cytokines, while SBD treatment markedly downregulated the mRNA levels of JNK, STAT3, IL1B, IL6, and TNF (Fig. 9B–F). As shown
in Fig. 10A and B, LPS stimulation led to a significant increase in ROS levels, with a noticeable enhancement in fluorescence intensity. However, SBD treatment significantly reduced
fluorescence intensity and ROS levels, indicating its potential anti-inflammatory and antioxidant effects. EFFECTS OF SBD ON JNK AXIS As illustrated in Fig. 11A–D, immunofluorescence, and
Western blot analysis demonstrated that LPS stimulation significantly increased JNK levels, leading to elevated fluorescence intensity and gene expression. After SBD treatment, both
fluorescence intensity and JNK were significantly reduced, suggesting that SBD effectively inhibits JNK activation. DISCUSSION Osteoarthritis (OA) is a common orthopedic disease that imposes
a substantial burden on society. Its prevalence increases with age, making OA an emerging global public health issue. Studies indicate that Traditional Chinese Medicine (TCM) shows
potential in treating various chronic diseases and tumors28,29,30,31. We employed a network pharmacology-based computational approach, combined with molecular docking, molecular dynamics
simulation, and experimental validation, to explore the potential mechanisms of SBD in the treatment of OA. Through an analysis of multiple databases, including GEO, we identified 114 active
chemical components and 113 intersecting targets, screening six active compounds with excellent binding affinities to core targets. Shinflavanone is an effective bone resorption inhibitor,
suggesting its potential to reverse OA pathology32. Gancaonin L and Gancaonin O, flavonoid compounds extracted from licorice, possess immunomodulatory and other biological activities, making
them promising candidates for OA treatment. Xambioona, a natural plant-derived compound, and its therapeutic mechanism in OA remain underexplored. Phaseol, a bioactive compound derived from
legumes, holds antioxidant, anti-inflammatory, and antidiabetic potential. Licorisoflavanone, an isoflavonoid compound, may have a bone-repairing effect. By utilizing Cytoscape software and
PPI network analysis, we identified four core targets from the compound-drug-component-disease-target network. The inflammatory cytokine Interleukin-1 beta(IL1β) induces an inflammatory
phenotype characterized by increased RANKL and matrix metalloproteinases (MMPs)33. Additionally, IL-1β enhances the production of inflammatory cytokines like Tumor Necrosis Factor (TNF) and
Interleukin-6(IL-6), exacerbating OA-related inflammation34. IL-1B inhibitors significantly reduce OA progression by preserving cartilage integrity and minimizing lesion size35. Elevated
IL-6 levels are associated with increased pain, stiffness, and radiographic severity36. Excessive IL-6 production negatively affects the synthesis of cartilage matrix proteins, accelerating
cartilage degradation37. Targeting IL-6 can alleviate its harmful effects on cartilage metabolism38. TNF influences various cellular processes that lead to cartilage degeneration and joint
pathology39. TNF plays a critical role in OA by activating signaling pathways such as NF-kappa B, MAPK, and PI3K/Akt, promoting inflammatory cell recruitment and chondrocyte degradation40.
TNF inhibitors have been shown to have the potential for inflammation relief and cartilage protection41. Activation of STAT3 is associated with the expression of IL-1β, IL-6, and TNF42,43.
The signal transducer and activator of transcription 3(STAT3) is involved in regulating chondrocyte apoptosis44,45. Therefore, simultaneously targeting these targets offers significant
potential for the treatment of OA. In the Gene Ontology (GO) analysis, Biological Processes (BP) were significantly associated with inflammation and cellular stress, including antioxidant
responses, TNF-mediated signaling, cellular responses to bacterial components and lipopolysaccharides, as well as the modulation of cell proliferation and apoptotic pathways. Cellular
Components (CC) were significantly enriched in regions such as lipid rafts, membrane microdomains, vesicle lumen, secretory granule lumen, and mitochondrial outer membrane, indicating that
these cellular components play crucial roles in inflammatory signaling and functional regulation. Molecular Functions (MF) involve ligand activity, DNA-binding transcription factor binding,
protein kinase regulatory activity, and receptor binding, which are likely associated with signal transduction and transcriptional regulation. GO analysis revealed the multidimensional roles
of SBD in cellular functions and processes. In the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, SBD may be multi-pathway synergistic in OA treatment. Key inflammation- and
immune-related pathways, including the AGE-RAGE pathway, TNF signaling pathway, IL-17 signaling pathway, and Toll-like receptor signaling pathway, indicate that SBD may exert therapeutic
effects on OA by regulating these signaling cascades. Reactive oxygen species (ROS) is a key driver of OA progression, playing a crucial role in inflammation and cartilage degradation in the
pathology of OA46. The AGE-RAGE axis enhances ROS generation while simultaneously inhibiting antioxidant enzyme activity, thereby weakening cellular defenses against oxidative stress47.
Increased oxidative stress leads to DNA damage and mitochondrial dysfunction in chondrocytes, exacerbating cartilage degeneration and inflammation48. The JNK axis is a key component in the
AGE-RAGE signaling pathway. JNK is considered a precursor of NF-κB. It not only exacerbates the inflammatory cascade but also promotes apoptosis by regulating the STAT349. Continued
activation of the JNK signaling pathway is implicated in cartilage degeneration and chronic synovial inflammation, making it a significant therapeutic target for OA50,51. Inhibiting JNK
expression helps alleviate the persistent cartilage degeneration and chronic inflammation in OA52. Inhibiting STAT3 activation is crucial in the progression of OA and cartilage damage53.
IL-17 signaling pathway is closely associated with OA progression. The crosstalk between the IL-17 pathway and the AGE-RAGE pathway may involve JNK, which activates NF-κB, triggering
inflammation and cartilage degradation, thereby further activating the IL-17 pathway54. Another study suggested the IL-17 axis as a novel therapeutic strategy for OA55. In OA animal models,
SPHK2 knockdown inhibited the IL-17 pathway and alleviated cartilage damage and synovial inflammation56. In rat chondrocytes, TNF has been shown to increase senescence markers and
glycosaminoglycan (GAG) degradation57. Clinical studies have demonstrated the potential of TNF inhibitors in treating arthritis58. Research indicates that IL-17 A and TNF synergistically
induce pro-inflammatory mediators in synovial fibroblasts, accelerating OA progression59. Overall, enrichment analysis suggests that SBD may effectively treat OA through multi-gene,
multi-pathway mechanisms. Using three algorithms (Degree Centrality, Maximum Clique Centrality, and Mean Nearest Neighbor Centrality), the top four core (IL1B, STAT3, IL6, TNF) genes were
identified. Furthermore, within the AGE-RAGE pathway, JNK is a common upstream target for the four core targets and ranks highly. Therefore, this study specifically explores the relationship
between the JNK axis in the AGE-RAGE pathway and OA. Molecular docking simulations suggest that the six key compounds (Shinflavanone, Gancaonin L, Xambioona, Phaseol, Gancaonin O, and
Licoisoflavanone) exhibit significant binding activity with the five target proteins (IL1B, STAT3, IL6, TNF, and JNK) under investigation. Shinflavanone exhibited the highest docking score
with JNK, demonstrating stable hydrogen bonds and strong van der Waals interactions. Shinflavanone_IL1B, Xambioona_IL6, Shinflavanone_STAT3, and Gancaonin L_TNF also showed favorable
interactions, primarily characterized by strong van der Waals forces and hydrophobic interactions. Molecular dynamics simulations further validated this finding by demonstrating the dynamic
stability of these interactions in solution. Root Mean Square Deviation (RMSD) and Root Mean Square Fluctuation (RMSF) analyses indicated that all ligand-protein complexes exhibited
relatively stable conformations, with Shinflavanone_JNK showing particularly stable behavior. The radius of gyration (Rg) and solvent-accessible surface area (SASA) analysis demonstrated the
stability of the binding interface throughout the simulation. The consistent number of hydrogen bonds (HB) maintained throughout the simulation also supports the stability of the complex
over time. The MM-PBSA method can quantitatively analyze the binding energy of ligand-protein complexes, offering unique advantages. MM-PBSA analysis further validated the dynamic stability
of these complexes. In conclusion, all complexes exhibited significant stability, providing strong theoretical support and data evidence for the development of new therapeutic approaches. To
validate the effects of SBD, we conducted in vitro experimental validation. The results indicate that SBD effectively scavenges ROS and inhibits the expression of IL-1β, IL-6, TNF-α, STAT3,
and JNK. This significantly highlights the potential of SBD in alleviating oxidative stress, exerting anti-inflammatory effects, and inhibiting cartilage degradation. In addition, both
immunofluorescence (IF) and Western blot (WB) experiments have demonstrated the importance of the JNK axis in the AGE-RAGE signaling pathway. Compared to conventional anti-inflammatory
therapies such as NSAIDs and biologics, SBD may exert a broader regulatory effect by simultaneously targeting multiple inflammatory mediators. In conclusion, Experimental validation further
supports the hypothesis that SBD alleviates OA by regulating the JNK axis within the AGE-RAGE signaling pathway, thereby disrupting the pathological loop in which JNK activation induces ROS
production, which in turn amplifies AGE-RAGE signaling, exacerbating inflammation and tissue damage. By breaking this vicious cycle, SBD may mitigate inflammatory responses and preserve
joint integrity. Future studies should explore its potential in combination with standard therapies to enhance efficacy and reduce adverse effects, offering a novel multi-targeted strategy
for OA management. CONCLUSION This study comprehensively explored the potential of SBD in the treatment of OA through network pharmacology, molecular docking, MD simulations, and
experimental validation. The results indicate that SBD may exert therapeutic effects on key inflammatory targets in OA through its key components, including Shinflavanone, Gancaonin L,
Xambioona, Phaseol, Gancaonin O, and Licoisoflavanone. Moreover, we validated that SBD alleviates oxidative stress, exhibits anti-inflammatory effects, and protects cartilage through the JNK
pathway. Overall, our findings confirm the reliability of computational techniques and highlight the potential of SBD as a therapeutic agent for OA (Fig. 12). DATA AVAILABILITY The novel
contributions of this study are detailed in the article and its supplementary materials. For any additional information, please contact the corresponding author. REFERENCES * Gelber, A. C.
Knee osteoarthritis. _Ann. Intern. Med._ 177. https://doi.org/10.7326/annals-24-01249 (2024). * Englund, M. Osteoarthritis, part of life or a curable disease? A bird’s-eye view. _J. Intern.
Med._ 293, 681–693. https://doi.org/10.1111/joim.13634 (2023). Article PubMed Google Scholar * Steinmetz, J. D.et al. Global, regional, and national burden of osteoarthritis, 1990–2020
and projections to 2050: A systematic analysis for the Global Burden of Disease Study 2021. _Lancet Rheumatol._ 5, e508–e522. https://doi.org/10.1016/S2665-9913(23)00163-7 (2023). * Zhang,
S. et al. Therapeutic potential of traditional Chinese medicine against osteoarthritis: targeting the Wnt signaling pathway. _Am. J. Chin. Med._ 52, 2021–2052.
https://doi.org/10.1142/s0192415x24500782 (2024). Article CAS PubMed Google Scholar * Zhou, G. et al. Research progress on the treatment of knee osteoarthritis combined with osteoporosis
by single-herb Chinese medicine and compound. _Front. Med. (Lausanne)_. 10, 1254086. https://doi.org/10.3389/fmed.2023.1254086 (2023). Article PubMed Google Scholar * Wang, L. et al.
Evaluation of the therapeutic effect of traditional Chinese medicine on osteoarthritis: A systematic review and meta-analysis. _Pain Res. Manag_. 2020, 5712187.
https://doi.org/10.1155/2020/5712187 (2020). * Wang, M. et al. Mechanism of traditional Chinese medicine in treating knee osteoarthritis. _J. Pain Res._ 13, 1421–1429.
https://doi.org/10.2147/jpr.S247827 (2020). Article PubMed PubMed Central Google Scholar * Xu, Z. et al. Exploring the mechanism of action of modified Simiao powder in the treatment of
osteoarthritis: an in-silico study. _Front. Med. (Lausanne)_. 11, 1422306. https://doi.org/10.3389/fmed.2024.1422306 (2024). Article PubMed Google Scholar * Padhee, S. et al.
Identification of the active constituents and molecular mechanism of Eulophia Nuda extract in the treatment of osteoarthritis by network pharmacology, molecular modeling, and experimental
assays. _Naunyn Schmiedebergs Arch. Pharmacol._ https://doi.org/10.1007/s00210-024-03459-z (2024). Article PubMed Google Scholar * Padhee, S. et al. Exploring the mechanism of action of
Vanda tessellate extract for the treatment of osteoarthritis through network pharmacology, molecular modeling, and experimental assays. _Heliyon_ 10, e35971.
https://doi.org/10.1016/j.heliyon.2024.e35971 (2024). Article CAS PubMed PubMed Central Google Scholar * Tanoli, Z., Schulman, A. & Aittokallio, T. Validation guidelines for
drug-target prediction methods. _Expert Opin. Drug Discov._ 1–15. https://doi.org/10.1080/17460441.2024.2430955 (2024). * Filipe, H. A. L. & Loura, L. M. S. Molecular dynamics
simulations: Advances and applications. _Molecules_ 27. https://doi.org/10.3390/molecules27072105 (2022). * Aranda-Garcia, D. et al. (2022). * Parise, A., Cresca, S. & Magistrato, A.
Molecular dynamics simulations for the structure-based drug design: targeting small-GTPases proteins. _Expert Opin. Drug Discov_. 19, 1259–1279. https://doi.org/10.1080/17460441.2024.2387856
(2024). Article CAS PubMed Google Scholar * Yang, M. et al. Molecular mechanism of Dang-Shen-Yu-Xing Decoction against Mycoplasma bovis pneumonia based on network pharmacology,
molecular docking, molecular dynamics simulations, and experimental verification. _Front. Vet. Sci._ 11, 1431233. https://doi.org/10.3389/fvets.2024.1431233 (2024). Article PubMed PubMed
Central Google Scholar * Li, X. et al. Combining network pharmacology, molecular docking, molecular dynamics simulation, and experimental verification to examine the efficacy and
immunoregulation mechanism of FHB granules on vitiligo. _Front. Immunol._ 14, 1194823. https://doi.org/10.3389/fimmu.2023.1194823 (2023). Article CAS PubMed PubMed Central Google Scholar
* Miao, J. et al. Exploring the therapeutic mechanisms of Yikang Decoction in polycystic ovary syndrome: an integration of GEO datasets, network pharmacology, and molecular dynamics
simulations. _Front. Med._ 11. https://doi.org/10.3389/fmed.2024.1455964 (2024). * Hua, Y. et al. Deciphering the Pharmacological mechanism of Radix astragali for allergic rhinitis through
network Pharmacology and experimental validation. _Sci. Rep._ 14, 29873. https://doi.org/10.1038/s41598-024-80101-1 (2024). Article CAS PubMed PubMed Central Google Scholar *
Klockmeier, K., Silva Ramos, E., Raskó, T., Martí Pastor, A. & Wanker, E. E. Schizophrenia risk candidate protein ZNF804A interacts with STAT2 and influences interferon-mediated gene
transcription in mammalian cells. _J. Mol. Biol._ 433, 167184. https://doi.org/10.1016/j.jmb.2021.167184 (2021). Article CAS PubMed Google Scholar * Choudhary, N., Choudhary, S., Kumar,
A. & Singh, V. Deciphering the multi-scale mechanisms of Tephrosia purpurea against polycystic ovarian syndrome (PCOS) and its major psychiatric comorbidities: studies from network
Pharmacological perspective. _Gene_ 773, 145385. https://doi.org/10.1016/j.gene.2020.145385 (2021). Article CAS PubMed Google Scholar * Yu, G., Wang, L. G., Han, Y. & He, Q. Y.
ClusterProfiler: an R package for comparing biological themes among gene clusters. _Omics_ 16, 284–287. https://doi.org/10.1089/omi.2011.0118 (2012). Article CAS PubMed PubMed Central
Google Scholar * Kanehisa, M., Furumichi, M., Sato, Y., Kawashima, M. & Ishiguro-Watanabe, M. KEGG for taxonomy-based analysis of pathways and genomes. _Nucleic Acids Res._ 51,
D587–d592. https://doi.org/10.1093/nar/gkac963 (2023). Article CAS PubMed Google Scholar * Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of Docking with a
new scoring function, efficient optimization, and multithreading. _J. Comput. Chem._ 31, 455–461. https://doi.org/10.1002/jcc.21334 (2010). Article CAS PubMed PubMed Central Google
Scholar * Morris, G. M. et al. AutoDock4 and AutoDockTools4: automated Docking with selective receptor flexibility. _J. Comput. Chem._ 30, 2785–2791. https://doi.org/10.1002/jcc.21256
(2009). Article CAS PubMed PubMed Central Google Scholar * Cheng, T., Li, X., Li, Y., Liu, Z. & Wang, R. Comparative assessment of scoring functions on a diverse test set. _J. Chem.
Inf. Model._ 49, 1079–1093. https://doi.org/10.1021/ci9000053 (2009). Article CAS PubMed Google Scholar * Yin, B., Bi, Y. M., Fan, G. J. & Xia, Y. Q. Molecular Mechanism of the
Effect of Huanglian Jiedu Decoction on Type 2 Diabetes Mellitus Based on Network Pharmacology and Molecular Docking. _J. Diabetes Res._, 5273914. https://doi.org/10.1155/2020/5273914 (2020).
* Pronk, S. et al. GROMACS 4.5: a high-throughput and highly parallel open source molecular simulation toolkit. _Bioinformatics_ 29, 845–854. https://doi.org/10.1093/bioinformatics/btt055
(2013). Article CAS PubMed PubMed Central Google Scholar * Liu, Y. et al. Traditional Chinese medicine in the treatment of chronic atrophic gastritis, precancerous lesions, and gastric
cancer. _J. Ethnopharmacol._ 337, 118812. https://doi.org/10.1016/j.jep.2024.118812 (2025). Article CAS PubMed Google Scholar * Wang, M. et al. Traditional Chinese medicine enhances the
effectiveness of immune checkpoint inhibitors in tumor treatment: A mechanism discussion. _J. Ethnopharmacol._ 338, 118955. https://doi.org/10.1016/j.jep.2024.118955 (2025). Article CAS
PubMed Google Scholar * Tan, P. et al. Application of omics technologies in studies on antitumor effects of traditional Chinese medicine. _Chin. Med._ 19, 123.
https://doi.org/10.1186/s13020-024-00995-x (2024). Article PubMed PubMed Central Google Scholar * Zhao, M., Che, Y., Gao, Y. & Zhang, X. Application of multi-omics in the study of
traditional Chinese medicine. _Front. Pharmacol._ 15, 1431862. https://doi.org/10.3389/fphar.2024.1431862 (2024). Article CAS PubMed PubMed Central Google Scholar * Suh, H., Lee, S.,
Kim, N., Han, J. & Kim, J. Syntheses of (+/-)-shin-flavanone and its structural analogs as potent inhibitors of bone resorption pits formation. _Bioorg. Med. Chem. Lett._ 9, 1433–1436.
https://doi.org/10.1016/s0960-894x(99)00212-7 (1999). Article CAS PubMed Google Scholar * Arra, M., Swarnkar, G., Alippe, Y., Mbalaviele, G. & Abu-Amer, Y. IκB-ζ signaling promotes
chondrocyte inflammatory phenotype, senescence, and erosive joint pathology. _Bone Res._ 10, 12. https://doi.org/10.1038/s41413-021-00183-9 (2022). Article CAS PubMed PubMed Central
Google Scholar * Mu, Y., Wang, L., Fu, L. & Li, Q. Knockdown of LMX1B suppressed cell apoptosis and inflammatory response in IL-1β-induced human osteoarthritis chondrocytes through
NF-κB and NLRP3 signal pathway. _Mediators Inflamm._ 1870579 (2022). https://doi.org/10.1155/2022/1870579 (2022). * Aman, Z. S. et al. Acute intervention with selective Interleukin-1
inhibitor therapy May reduce the progression of posttraumatic osteoarthritis of the knee: A systematic review of current evidence. _Arthroscopy_ 38, 2543–2556.
https://doi.org/10.1016/j.arthro.2022.02.009 (2022). Article PubMed Google Scholar * Ahmed, R., Soliman, N. & Elwan, S. Relationships between serum Interleukin-6, radiographic
severity a WOMAC index in patients with primary knee osteoarthritis. (2023). * Eitner, A. et al. Importance of IL-6 trans-signaling and high autocrine IL-6 production in human Osteoarthritic
chondrocyte metabolism. _Osteoarthr. Cartil._ 32, 561–573. https://doi.org/10.1016/j.joca.2024.02.006 (2024). Article Google Scholar * Shanshal, A., Hisham, R. & Hussain, S. Targeting
IL-6 signaling pathways for musculoskeletal disorders treatment: Risks and benefits. _Al-Rafidain J. Med. Sci._ 4, 34–43. https://doi.org/10.54133/ajms.v4i.101 (2023). * Fedulichev, P. N.
The role of immune factors in the etiopathogenesis of osteoarthritis. _Сибирский Научный Медицинский Журнал_ (2023). * Guo, Y. et al. Kongensin A targeting PI3K attenuates
inflammation-induced osteoarthritis by modulating macrophage polarization and alleviating inflammatory signaling. _Int. Immunopharmacol._ 142, 112948.
https://doi.org/10.1016/j.intimp.2024.112948 (2024). Article CAS PubMed Google Scholar * Valerio, M. S. et al. Effect of targeted cytokine Inhibition on progression of post-traumatic
osteoarthritis following Intra-Articular fracture. _Int. J. Mol. Sci._ 24. https://doi.org/10.3390/ijms241713606 (2023). * Lu, X. et al. Selective STAT3 inhibitor STX-0119 alleviates
osteoarthritis progression by modulating the STAT3/PPARγ signaling pathway. _Biochem. Pharmacol._ 227, 116420. https://doi.org/10.1016/j.bcp.2024.116420 (2024). Article CAS PubMed Google
Scholar * Moon, J. et al. Small heterodimer partner-interacting leucine zipper protein suppresses pain and cartilage destruction in an osteoarthritis model by modulating the AMPK/STAT3
signaling pathway. _Arthritis Res. Ther._ 26, 199. https://doi.org/10.1186/s13075-024-03417-3 (2024). Article CAS PubMed PubMed Central Google Scholar * Li, J., Yin, Z., Huang, B., Xu,
K. & Su, J. Stat3 signaling pathway: A future therapeutic target for Bone-Related diseases. _Front. Pharmacol._ 13, 897539. https://doi.org/10.3389/fphar.2022.897539 (2022). Article CAS
PubMed PubMed Central Google Scholar * Kaneko, Y. et al. The Stat3 inhibitor F0648-0027 is a potential therapeutic against rheumatoid arthritis. _Biochem. Biophys. Res. Commun._ 636,
133–140. https://doi.org/10.1016/j.bbrc.2022.10.106 (2022). Article CAS PubMed Google Scholar * Fang, D. et al. Ros-responsive nanocomposite scaffolds for sustained releasing puerarin to
achieve chondroprotection in OA rats. _Mater. Design_. 233, 112214. https://doi.org/10.1016/j.matdes.2023.112214 (2023). Article CAS Google Scholar * Bhattacharya, R., Alam, M. R.,
Kamal, M. A., Seo, K. J. & Singh, L. R. AGE-RAGE axis culminates into multiple pathogenic processes: a central road to neurodegeneration. _Front. Mol. Neurosci._ 16, 1155175.
https://doi.org/10.3389/fnmol.2023.1155175 (2023). Article CAS PubMed PubMed Central Google Scholar * Coryell, P. R., Diekman, B. O. & Loeser, R. F. Mechanisms and therapeutic
implications of cellular senescence in osteoarthritis. _Nat. Rev. Rheumatol._ 17, 47–57. https://doi.org/10.1038/s41584-020-00533-7 (2021). Article PubMed Google Scholar * Nadel, G.,
Maik-Rachline, G. & Seger, R. J. N. K. Cascade-Induced apoptosis-A unique role in GqPCR signaling. _Int. J. Mol. Sci._ 24. https://doi.org/10.3390/ijms241713527 (2023). * 50 Chen, B.,
Ning, K., Sun, M. L. & Zhang, X. A. Regulation and therapy, the role of JAK2/STAT3 signaling pathway in OA: a systematic review. _Cell. Commun. Signal._ 21, 67.
https://doi.org/10.1186/s12964-023-01094-4 (2023). Article PubMed PubMed Central Google Scholar * Zhou, Q. et al. The potential roles of JAK/STAT signaling in the progression of
osteoarthritis. _Front. Endocrinol. (Lausanne)_. 13, 1069057. https://doi.org/10.3389/fendo.2022.1069057 (2022). Article PubMed Google Scholar * 52 Qian, Z. et al. AFK-PD alleviated
osteoarthritis progression by chondroprotective and anti-inflammatory activity. _Front. Pharmacol._ 15, 1439678. https://doi.org/10.3389/fphar.2024.1439678 (2024). Article CAS PubMed
PubMed Central Google Scholar * Li, J. et al. Endothelial Stat3 activation promotes osteoarthritis development. _Cell. Prolif._ 56, e13518. https://doi.org/10.1111/cpr.13518 (2023).
Article CAS PubMed PubMed Central Google Scholar * Xiao, J. et al. IL-17 in osteoarthritis: A narrative review. _Open. Life Sci._ 18, 20220747. https://doi.org/10.1515/biol-2022-0747
(2023). Article CAS PubMed PubMed Central Google Scholar * Hsieh, S. L. et al. MCP-1 controls IL-17-promoted monocyte migration and M1 polarization in osteoarthritis. _Int.
Immunopharmacol._ 132, 112016. https://doi.org/10.1016/j.intimp.2024.112016 (2024). Article CAS PubMed Google Scholar * Zheng, J. et al. SPHK2 knockdown inhibits the proliferation and
migration of Fibroblast-Like synoviocytes through the IL-17 signaling pathway in osteoarthritis. _J. Inflamm. Res._ 17, 7221–7234. https://doi.org/10.2147/jir.S476077 (2024). Article PubMed
PubMed Central Google Scholar * Yagi, M., Endo, K., Komori, K. & Sekiya, I. Comparison of the effects of oxidative and inflammatory stresses on rat chondrocyte senescence. _Sci.
Rep._ 13, 7697. https://doi.org/10.1038/s41598-023-34825-1 (2023). Article ADS CAS PubMed PubMed Central Google Scholar * Wang, W. et al. Serum IL-40 increases in patients with
rheumatoid arthritis and correlates with some clinical characteristics and comorbidities. _Sci. Rep._ 14, 28945. https://doi.org/10.1038/s41598-024-80104-y (2024). Article CAS PubMed
PubMed Central Google Scholar * Kouri, V. P. et al. IL-17A and TNF synergistically drive the expression of Proinflammatory mediators in synovial fibroblasts via IκBζ-dependent induction of
ELF3. _Rheumatol. (Oxford)_. 62, 872–885. https://doi.org/10.1093/rheumatology/keac385 (2023). Article Google Scholar Download references ACKNOWLEDGEMENTS The authors sincerely appreciate
the significant contributions of both the researchers and the participants who played an essential role in this study. FUNDING This study received financial support from the Science and
Technology Program Project of Chongzuo City (Grant No. Chong Ke 2023QN035568). AUTHOR INFORMATION Author notes * These authors contributed equally: Zeyu Huang and Xiaohong Jiang. AUTHORS AND
AFFILIATIONS * Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530001, China Zeyu Huang, Xiaohong Jiang, Wei Xie, Kuicheng Wei & Dehuai Liu * Big
Data Technology Development Division, Guangxi Zhuang Autonomous Region Information Center, Nanning, 530221, China Lianlian Zhong * Department of Epidemiology and Health Statistics, School
of Public Health, Guangxi Medical University, Nanning, 530021, China Xiaohong Jiang * Department of Orthopedics, Affiliated Hospital of Guilin Medical University, Guilin, 541000, China
Lerong Yang Authors * Zeyu Huang View author publications You can also search for this author inPubMed Google Scholar * Xiaohong Jiang View author publications You can also search for this
author inPubMed Google Scholar * Wei Xie View author publications You can also search for this author inPubMed Google Scholar * Kuicheng Wei View author publications You can also search for
this author inPubMed Google Scholar * Lerong Yang View author publications You can also search for this author inPubMed Google Scholar * Dehuai Liu View author publications You can also
search for this author inPubMed Google Scholar * Lianlian Zhong View author publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS Conceptualization and
methodology, Zeyu Huang and Kuicheng Wei; software, Lianlian Zhong, Zeyu Huang; validation, Zeyu Huang, Xiaohong Jiang and Dehuai Liu; formal analysis, Dehuai Liu, Zeyu Huang, Wei Xie and
Kuicheng Wei; investigation and data curation, Zeyu Huang, Wei Xie and Dehuai Liu; writing—original draft preparation, Zeyu Huang and Lianlian Zhong; writing—review and editing, Xiaohong
Jiang, Zeyu Huang, Lerong Yang, Dehuai Liu; visualization, Zeyu Huang, Xiaohong Jiang and Lianlian Zhong; resources, supervision, project administration and funding acquisition, Dehuai Liu.
All authors have read and agreed to the published version of the manuscript. CORRESPONDING AUTHORS Correspondence to Dehuai Liu or Lianlian Zhong. ETHICS DECLARATIONS COMPETING INTERESTS The
authors declare no competing interests. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional
affiliations. ELECTRONIC SUPPLEMENTARY MATERIAL Below is the link to the electronic supplementary material. SUPPLEMENTARY MATERIAL 1 SUPPLEMENTARY MATERIAL 2 SUPPLEMENTARY MATERIAL 3
SUPPLEMENTARY MATERIAL 4 SUPPLEMENTARY MATERIAL 5 SUPPLEMENTARY MATERIAL 6 RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons
Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission
under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons
licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by
statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit
http://creativecommons.org/licenses/by-nc-nd/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Huang, Z., Jiang, X., Xie, W. _et al._ Mechanistic insights into Sanbi
Decoction for osteoarthritis treatment based on network pharmacology and experimental validation. _Sci Rep_ 15, 18707 (2025). https://doi.org/10.1038/s41598-025-99055-z Download citation *
Received: 26 January 2025 * Accepted: 16 April 2025 * Published: 28 May 2025 * DOI: https://doi.org/10.1038/s41598-025-99055-z SHARE THIS ARTICLE Anyone you share the following link with
will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt
content-sharing initiative KEYWORDS * Sanbi Decoction * Osteoarthritis * Network pharmacology * Molecular docking * Molecular dynamics simulations.