Abstract Background Periodontitis is the primary cause of tooth loss in adults and is associated with cardiovascular disorders and type 2 diabetes, etc., significantly impairing patients’ quality of life. Salvia miltiorrhiza(S. miltiorrhiza), a traditional Chinese medicine, possesses properties such as anti-inflammatory and antioxidative effects and has potential in the treatment of periodontitis; however, its mechanism of action remains unclear. The aim of this study was to investigate the therapeutic mechanism of S. miltiorrhiza in periodontitis using an integrated approach combining network pharmacology, molecular docking, molecular dynamics simulations, and experimental validation. Methods The active components and target genes of Salvia miltiorrhiza were collected from the TCMSP and SwissTargetPrediction databases, while the target genes of periodontitis were obtained from the GeneCards, OMIM, Disgenet, and DrugBank databases. The intersection of these targets was identified using jvenn, followed by network visualization using STRING and Cytoscape 3.10.2 software. CytoNCA plug-ins were used to calculate node scores and identify hub genes. The DAVID database was used to conduct Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Molecular docking and molecular dynamics simulation were employed to evaluate the affinity and stability between key active compounds of S.miltiorrhiza and the hub genes. A RAW264.7 cell model induced by Pg-LPS (Porphyromonas gingivalis-lipopolysaccharide) was established. A CCK-8 assay was used to determine the effects of S.miltiorrhiza on the viability of RAW264.7 cells, thereby screening for appropriate drug concentrations. The Griess method was used to test the effect of S. miltiorrhiza on nitric oxide (NO) in cells. The mRNA expression levels of inflammation-related factors were detected by RT-qPCR. Results A total of 65 compounds from S. miltiorrhiza and 132 corresponding target genes were identified, along with 1900 periodontitis-related target genes. The intersection of these targets revealed 60 common targets. PPI network analysis revealed that S. miltiorrhiza may alleviate periodontitis by modulating key genes, including IL-6, BCL2, STAT3, TNF, TP53, CASP3, and MMP9. Molecular docking indicated strong binding affinities between the bioactive compounds in S. miltiorrhiza and these critical targets. Functional enrichment analysis suggested that the anti-inflammatory action of S. miltiorrhiza in periodontitis may involve the regulation of pathways such as AGE-RAGE, TNF and PI3k/AKT1 pathways. The results of cell experiments revealed that S. miltiorrhiza could treat and prevent periodontitis by inhibiting NO production and regulating the mRNA expression of inflammatory factors, including IL-1β, TNF, IL-6, and IL-10. Conclusion S. miltiorrhiza exerts therapeutic effects on periodontitis via multiple components, targets and pathways, which provides a sufficient theoretical and practical basis for the further study of S. miltiorrhiza in the treatment of periodontitis. Keywords: Salvia miltiorrhiza(SM), Periodontitis, Network pharmacology, Molecular docking method, Molecular dynamics simulation, Inflammation Introduction Periodontitis, the sixth most prevalent global disease, affects 11.2% of the worldwide population [[32]1]. This persistent inflammatory condition is primarily caused by plaque accumulation. Furthermore, its development is influenced by several additional factors, including bacterial infections, irritation from inadequate dental restorations, endocrine abnormalities, genetic predisposition, and environmental influences [[33]2].The fundamental concept underlying the treatment of periodontitis is the use of mechanical techniques to eliminate calculus and plaque from teeth [[34]3]. Nevertheless, in practical scenarios, dental surfaces that are inaccessible to equipment exist, and mechanical techniques are unable to completely eliminate all surface bacteria [[35]4]. Consequently, antibiotics are frequently administered in conjunction with mechanical therapy. Notably, prolonged antibiotic use is associated with bacterial resistance and other adverse effects [[36]5]. Traditional Chinese medicine (TCM) has been widely employed in the treatment of periodontitis due to its minimal side effects, substantial efficacy, and low propensity to induce drug resistance [[37]6]. For nearly two millennia, S. miltiorrhiza, an herb widely used in traditional Chinese medicinal formulations, has been employed in China to address various clinical conditions. Research has demonstrated that S. miltiorrhiza has potential health benefits in terms of anti-inflammatory, anticoagulant, and antioxidant activities, thereby offering new possibilities for the treatment and prevention of periodontitis [[38]7]. While several studies have shown the multisystemic pharmacological effects of S. miltiorrhiza, the domain of measurement Chinese medicine is insufficiently broad to evaluate the pharmacological effects of S. miltiorrhiza from a singular system. Network pharmacology, an innovative research paradigm, focuses on integrating disease networks, gene networks, and therapeutic target networks [[39]8]. Molecular docking is a methodology that employs stoichiometry to simulate the geometric arrangement of molecules and intermolecular forces, facilitating the identification of intermolecular interactions and the prediction of receptor-ligand interaction complex configurations [[40]9]. Molecular dynamics employs computational methods to solve molecular motion equations, simulating the structure and properties of molecular systems [[41]10]. Recently, accumulating evidence has demonstrated that computer-aided drug design approaches can predict the active components, targets, and action mechanisms of natural compounds; this not only significantly shortens the timeline and reduces costs in new drug development but also opens novel research frontiers for natural product exploitation [[42]11]. The purpose of this study was to explore the potential targets and signalling pathways of S. miltiorrhiza in treating periodontitis through network pharmacology, molecular docking, and molecular dynamics simulation, providing a robust theoretical foundation for future experimental investigations and clinical applications of this herb. Additionally, an in vitro cell model was established to assess the regulatory effect of S. miltiorrhiza on inflammatory cytokine secretion by macrophages stimulated with Pg-LPS, offering theoretical support for its development as a therapeutic agent for periodontitis.All precise procedures of the method are illustrated in Fig. [43]1. Fig. 1. [44]Fig. 1 [45]Open in a new tab Workflow of a pharmacology-based approach to investigate the therapeutic mechanisms of S. miltiorrhiza on periodontitis Materials and methods Evaluation of the active components and targets of S. miltiorrhiza To identify the main components of S. miltiorrhiza, we searched the Traditional Chinese Medicine Systematic Pharmacology Database and Analysis Platform (TCMSP). The criteria for screening active components in S. miltiorrhiza were formulated based on the pharmacokinetic (ADME) properties of drugs in traditional Chinese medicine. The goal parameters were a bioavailability (OB) of at least 30% and a drug-like property (DL) of at least 0.18. Targets associated with the candidate bioactive compounds were identified via the SwissTargetPrediction database. The standard gene names of the relevant targets were identified by checking the acquired target names in the UniProt database. (The website of the public databases was shown in Table [46]1) Table 1. Software and database for network Pharmacology Name Website TCMSP [47]https://tcmspw.com/tcmsp.php SwissTargetPrediction [48]http://www.swisstargetprediction.ch/ Genecard [49]https://www.genecards.org/ Omim [50]https://www.omim.org/ Disgenet [51]http://DisGeNET.org Drugbank [52]https://go.drugbank.com/ Venny 2.1.0 [53]https://bioinfogp.cnb.csic.es/tools/venny/ STRING [54]https://cn.string-db.org/ DAVID [55]https://david.ncifcrf.gov/ Bioinformatics platform [56]http://www.bioinformatics.com.cn/ PubChem [57]https://pubchem.ncbi.nlm.nih.gov/ PDB [58]https://www.rcsb.org/ Uniprot [59]https://www.uniprot.org/ [60]Open in a new tab Target prediction of S. miltiorrhiza in the treatment of periodontitis Using the term “periodontitis” as the search term, we conducted a search for the databases Genecard, Omim, Disgenet, and DrugBank to ascertain the objective of periodontitis-related disorders. The aims were subsequently combined and stripped of their emphasis. A crossover network of targets of active ingredients in S. miltiorrhiza and periodontitis disease-related targets was constructed via Venny 2.1.0 to screen the intersection targets used for the following analyses. (The website of the public databases and analysis tool was shown in Table [61]1). Development and examination of PPI networks Through the use of the STRING database, the common target genes for periodontitis in S. miltiorrhiza were identified. The construction of the PPI network required the application of a filter that had a confidence level that was greater than 0.7. Following elimination of the free nodes, the nodes were imported into Cytoscape 3.10.2 for visual analysis. The network evaluation study employed CyoNCA to systematically assess the effectiveness of S. miltiorrhiza treatment for periodontitis based on the node degree and neutrality value. GO function and KEGG enrichment analysis Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed via DAVID to determine the role of Astragali radix in biological processes, molecular functions, cellular components and signalling pathways in the treatment of periodontitis. The results were visualized on a bioinformatics platform. Statistical significance was observed at p < 0.05, with a smaller p value indicating greater enrichment and a larger count indicating that more genes were enriched (Table [62]1). Map of the relationship between S. miltiorrhiza-active ingredients and target networks To acquire a better understanding of the S. miltiorrhiza-active ingredient-target network, we removed redundant items from the Excel spreadsheet containing the active ingredient and its related target with S. miltiorrhiza. All tables were then loaded into the Cytoscape 3.10.2 program. Correlation analysis of drugs with disease targets To generate the original network, the encouraging nodes were removed, and the intersecting drug and disease targets (2.2) were added to the String database. The PPIs were then analysed and correlated, and the intersecting nodes were then removed. Molecular docking analysis Molecular docking was performed to validate the target prediction results by docking selected active ingredients with hub genes. The bioactive components and protein structures of the hub genes were retrieved from the PubChem and PDB databases.The AutoDockTools1.1.2 software was utilized in order to establish a docking relationship between the component elements of the medication and the target proteins. Subsequently, PyMOL software was used to visualize the results. Molecular dynamics simulations Molecular dynamics simulations were performed using Amber 24 (San Francisco, CA, USA) to examine the interaction mechanisms between molecular ligands and protein receptors and to assess binding stability. Simulation conditions were set at a constant temperature of 300 K and atmospheric pressure (1 Bar). The Amber99sb-ildn force field was applied, with water molecules as the solvent (Tip3p model). The simulation system’s total charge was neutralized by adding appropriate Na + ions. Energy minimization was conducted via the steepest descent method, followed by 100,000-step equilibrations under isothermal-isovolumic (NVT) and isothermal-isobaric (NPT) ensembles, respectively. The coupling constant was 0.1 ps, and each equilibration lasted 100 ps.Subsequently, a 100-ns free molecular dynamics simulation was executed, comprising 5 million steps with a 2-fs time step. Post-simulation, trajectory analysis was performed using the software’s built-in tools to calculate root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), and protein radius of gyration for each amino acid. These analyses were combined with free energy calculations (MMGBSA) and free energy landscape data. In vitro experiment Chemicals and materials S. miltiorrhiza was purchased from Tongrentang Pharmacy (Beijing, China). Foetal bovine serum (FBS) was obtained from Oricell (USA) and used as a supplement for cell culture. Dulbecco’s Modified Eagle Medium (DMEM), sourced from Gibco (USA), served as the primary culture medium. Phosphate-buffered saline (PBS), procured from Biosharp (China), was used for washing and dilution. For the cell viability assays, the CCK-8 kit was supplied by Biyuntian (China). P. gingivalis lipopolysaccharide (P. gingivalis LPS), a key experimental agent, was purchased from InvivoGen (USA). Additionally, chloroform, isopropyl alcohol, and anhydrous ethanol, which are essential for RNA extraction and purification, were obtained from Guangdong Guanghua Technology (China). DEPC-treated water, which was used to prevent RNA degradation, was obtained from Blue Sky (China). To extract RNA, the TRIzol-based TRleasyTM Total RNA Extraction reagent manufactured by Yisheng Biology in China was utilized. To perform complementary DNA (cDNA) synthesis, a premix kit from Yisheng Biology (China) was used. Additionally, quantitative polymerase chain reaction (qPCR) analysis was conducted using the qPCR SYBR Green Kit, which was also provided by Yisheng Biology (China). Preparation of S. miltiorrhiza extract The extraction of S. miltiorrhiza was followed a literature-based protocol: the material was extracted twice with water at 80°C for 120 min per cycle. The resulting aqueous extracts were combined, filtered, and concentrated under vacuum at 60°C to a relative density of 1.18 to 1.22. Ethanol was then added to achieve 70% (v/v) alcohol concentration, and the solution was allowed to stand for 12 h. The supernatant was collected, after which ethanol was recovered under reduced pressure. The remaining solution was concentrated to a thick paste and dried in a vacuum oven to yield the S. miltiorrhiza extract (yield: 10%) [[63]12]. Methods of cell treatment and culture In this particular investigation, the mouse macrophage line RAW264.7 was utilized. This particular line was obtained from the Laboratory of Oral Biology at Jilin University. To cultivate the cells, Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% foetal bovine serum (FBS) and 1% cyocyanin was added throughout the growth process. For optimal development conditions, cultures were maintained in an incubator that was humidified and maintained at 37 °C with 5% carbon dioxide. To maintain cell viability and proliferation, RAW264.7 macrophages were subcultured every two days. Cell counting kit 8 (CCK-8) was utilized to assess cell viability To seed RAW264.7 cells (murine macrophage line, catalogue number: TCM13) into 96-well plates, a density of 0.5 × 10⁴ cells was added to each well. This was performed at a specific concentration. Following the adherence of the cells to the surface of the plate, the culture media was changed to complete media that contained S. miltiorrhiza extract, and the cells were then incubated for approximately twenty-four hours. Subsequently, 100 µL of DMEM was added to each well, into which 10 µL of CCK-8 reagent was added. The plates were incubated for one hour in 5% CO₂, and 450 nm absorbance was measured. The absorbance measurements were used to create a cell viability curve to identify the best S. miltiorrhiza extract concentration for future research. Relationship between S. miltiorrhiza and the generation of NO in RAW264.7 cells For the purpose of seeding the RAW264.7 macrophage line onto 6-well plates, a total of 2 × 10⁵ cells were individually added to each well. The amount of cell suspension added to each well was 2 mL. After an initial incubation period of twenty-four hours, which was designed to allow the cells to attach to one another and become more stable, the cells were then treated with specialized medium that corresponded to the experimental groups. These groups included the blank control group, the LPS stimulation group (treated with lipopolysaccharide alone), and three experimental groups treated with low, medium, or high concentrations of S. miltiorrhiza extract. After 24 h, 50 µL of the supernatant was collected from each well and transferred to a 96-well plate. Subsequently, 50 µL of Griess Reagents I and II were added to each well at room temperature. After an incubation period of three minutes, the absorbance of each well was measured at 540 nm using an enzyme-linked immunosorbent assay reader. The absorbance of each well was measured. Sodium nitrite (NaNO₂) was utilized to create a standard curve, which facilitated the determination of nitric oxide (NO) content in the growth medium for each experimental group. Real-time quantitative fluorescence PCR (qRT-PCR) was used to assess the expression levels of TNF-α, IL-6, IL-1β and IL-10 mRNA RAW264.7 cells were seeded onto 6-well plates at a density of 2 × 10⁵ cells per well when the plates were prepared. Following the adherence of the cells to the surface of the plate, the culture media was changed to complete media that contained different quantities of lipopolysaccharide (LPS) and S. miltiorrhiza extract. TRIzol was used to extract total RNA after 24 h of therapy. A NanoDrop 2000 spectrophotometer was used to measure the RNA concentration, and a reverse transcription kit was used to produce cDNA. Quantitative real-time polymerase chain reaction (qRT-PCR) was carried out to evaluate the mRNA expression levels of TNF-α, IL-6, IL-1β and IL-10. The quantification of the expression levels of these cytokines was carried out by applying the 2^-ΔΔCt approach, with β-actin serving as the internal reference gene for standardization.The primer sequence is displayed in Table [64]2. Table 2. Primer sequences of Real-time PCR Gene Sequence of Primers (5ʹ→3ʹ) β-actin F: GGAGATTACTGCCCTGGCTCCTA R: GACTCATCGTACTCCTGCTTGCTG IL-6 F: GGAGCCCACCAAGAACGATA R: ACCAGCATCAGTCCCAAGAA TNF-α F: CTCATGCACCACCATCAAGG R: ACCTGACCACTCTCCCTTTG IL-1β F: ATGAAGGGCTGCTTCCAAAC R: TCTCCACAGCCACAATGAGT IL-10 F: GCTCTTACTGACTGGCATGAG R: CGCAGCTCTAGGAGCATGTG [65]Open in a new tab Statistical analyses Data analysis and graphing of the experimental data were performed using GraphPad Prism 9.0 software. The results of the above - mentioned in vitro experiments were replicated three times, and all results were presented as mean ± standard deviation (± SD). For comparisons between groups, if the data conformed to the normal distribution and passed the homogeneity of variance test, one - way analysis of variance (ANOVA) was employed. Statistical significance was defined as p < 0.05. Results Evaluation of S. miltiorrhiza active components and action targets A total of 65 main components of S. miltiorrhiza were found through TCMSP database (Table [66]3)0.132 active metabolite-related targets of S.miltiorrhiza were obtained. Table 3. Active components of S. miltiorrhiza MOL Compound OB DL MOL001601 1,2,5,6-tetrahydrotanshinone 38.74 0.35 MOL001659 Poriferasterol 43.82 0.75 MOL001771 poriferast-5-en-3beta-ol 36.91 0.75 MOL001942 isoimperatorin 45.46 0.22 MOL002222 sugiol 36.11 0.27 MOL002651 Dehydrotanshinone II A 43.76 0.4 MOL002776 Baicalin 40.12 0.75 MOL000569 digallate 61.84 0.25 MOL000006 luteolin 36.16 0.24 MOL006824 α-amyrin 39.51 0.76 MOL007036 5,6-dihydroxy-7-isopropyl-1 33.76 0.28 MOL007041 2-isopropyl-8-methylphenanthrene-3,4-dione 40.86 0.22 MOL007045 3α-hydroxytanshinoneIIa 44.92 0.44 MOL007048 acrylic acid 48.24 0.31 MOL007049 4-methylenemiltirone4 34.34 0.22 MOL007050 2-(4-hydroxy-3-methoxyphenyl)-5-(3-hydroxypropyl)-7-methoxy-3-benzofura ncarboxaldehyde 62.78 0.39 MOL007051 6-o-syringyl-8-o-acetyl shanzhiside methyl ester 46.69 0.71 MOL007058 formyltanshinone 73.44 0.41 MOL007059 3-beta-Hydroxymethyllenetanshiquinone 32.16 0.4 MOL007061 Methylenetanshinquinone 37.07 0.36 MOL007063 przewalskin a 37.1 0.64 MOL007064 przewalskin b 110.32 0.43 MOL007068 Przewaquinone B 62.24 0.41 MOL007069 przewaquinone c 55.74 0.4 MOL007070 (6 S,7R)-6,7-dihydroxy-1,6-dimethyl-8,9-dihydro-7 H-naphtho[8,7-g]benzo furan-10,11-dione 41.31 0.45 MOL007071 przewaquinone f 40.3 0.45 MOL007071 przewaquinone f 43.67 0.2 MOL007071 przewaquinone f 52.47 0.45 MOL007071 przewaquinone f 57.95 0.55 MOL007071 przewaquinone f 56.96 0.52 MOL007071 przewaquinone f 30.38 0.37 MOL007088 cryptotanshinone 52.34 0.39 MOL007093 dan-shexinkum d 38.88 0.55 MOL007094 danshenspiroketallactone 50.43 0.3 MOL007098 deoxyneocryptotanshinone 49.4 0.28 MOL007100 dihydrotanshinlactone 38.68 0.32 MOL007101 dihydrotanshinoneI 45.04 0.36 MOL007105 epidanshenspiroketallactone 68.27 0.3 MOL007107 [67]C09092 36.06 0.24 MOL007108 isocryptotanshi-none 54.98 0.39 MOL007111 Isotanshinone II 49.91 0.39 MOL007115 manool 45.04 0.2 MOL007118 microstegiol 39.61 0.27 [68]Open in a new tab Acquisition of gene targets for periodontitis Genetic targets for periodontitis were identified in the GeneCards database. These genes were subsequently refined using the median relevance score planting, resulting in a final collection of 1626 gene targets related to periodontitis. By merging and downsizing the gene targets from the OMIM database, an additional 8 gene targets related to periodontitis were identified. Furthermore, 682 additional gene targets related to periodontitis were discovered in the DisGeNET database. An analysis of the DrugBank database yielded twenty-eight gene targets associated with periodontitis. When the gene targets related to periodontitis were extracted from the four databases, they were categorized and minimized once the extraction process was complete. Finally, the 1900 gene targets related to periodontitis were identified. Identification of the intersection of periodontitis gene targets and S. miltiorrhiza active ingredient targets of S. miltiorrhiza Through the online analysis tool Venn diagram, the predicted targets of S. miltiorrhiza and periodontitis were analysed, and the overlapping targets obtained were used as the core targets of S. miltiorrhiza in the prevention of periodontitis. As shown in Fig. [69]2A, there were 60 intersecting genes between S. miltiorrhiza and periodontitis. Fig. 2. Fig. 2 [70]Open in a new tab The network pharmacological analysis of S. miltiorrhiza and periodontitis (A): Venn diagram of targets of active components of S. miltiorrhiza and periodontitis treatment-related targets; B: Network of PPI C: Potential core targets of S. miltiorrhiza in the treatment of periodontitis D: S. miltiorrhiza component-target-pathway network for periodontitis therapy. E: GO pathway enrichment analysis; F: KEGG pathway enrichment analysis Construction and analysis of PPI networks and acquisition of core targets Following the establishment of a high level of confidence and burying of the individual targets, the overlapping targets were loaded into the STRING database (Fig. [71]2B). Additionally, a PPI network map of the core targets was constructed via Cytoscape 3.10.2, and the top 7 targets were selected according to their MCC values. IL-6, BCL2, STAT3, TNF, TP53, CASP3, and MMP9 are the core targets of S. miltiorrhiza in preventing periodontitis (Fig. [72]2C). Relationship diagrams for the construction of S.miltiorrhiza active ingredient-target networks The 65 active chemicals identified in Sect. 3.1 were amalgamated and subsequently de-emphasized, resulting in 132 probable action targets for S. miltiorrhiza active compounds, which were determined by predicting the relevant targets via the Swiss Target Prediction database. From the 132 possible action targets of S. miltiorrhiza, 18 active compounds associated with these targets were identified. The active ingredient-target network graph had 151 nodes and 116 edges (Fig. [73]2D). The 132 nodes illustrated as green circles in the graph denote the target genes of S. miltiorrhiza for periodontitis treatment, whereas the remaining 18 nodes indicate the active components of S. miltiorrhiza linked to these targets. S. miltiorrhiza was produced via Cytoscape. The network diagram shows S. miltiorrhiza, active compounds, and target interactions. GO and KEGG pathway enrichment analysis The DAVID database indicates that GO and KEGG analyses of the prospective therapeutic targets of Salvia miltiorrhiza in periodontitis have been conducted. According to the findings of the GO analysis, the majority of the factors that were included in MF were gene expressiona and apoptotic, etc. In contrast, the majority of the factors that were included in the CC were macromolecular complex and nucleoplasm, etc. However, the majority of the factors associated with BP were enzyme binding, identical protein binding and protein binding, among others (Fig. [74]2E). According to KEGG data, the primary targets of S. miltiorrhiza in periodontitis include the AGE-RAGE, TNF, PI3K-Akt, HIF-1, and IL-17 signalling pathways in diabetic complications (Fig. [75]2F). These findings indicate that S. miltiorrhiza exerts its therapeutic effect on periodontitis through a multifaceted and multichannel mechanism. Molecular docking results and visualization Combining the degree value of protein interactions of key targets in the PPI network analysis, the network degree analysis of “drug-active ingredient-target-disease network”, literature review [[76]13], and the core active ingredients Baicalin (CID:64982), Digallate (CID: 54711004), and Luteolin (CID: 5280445), Poriferasterol (CID: 5281330) and Sugiol (CID: 94162) were selected for docking with the core target proteins AKT1 (ID: 1H10), CASP3 (2CJY), IL-6 (ID: 1ITV), MMP-9 (ID: 1ITV), STAT3 (ID: 5AX3), TNF (ID: 5M2J) and TP53 (ID: 3LGF). The molecular docking results revealed that the binding energy of these active ingredients docked with key target proteins was less than − 5 kcal/mol (Fig. [77]3). Fig. 3. [78]Fig. 3 [79]Open in a new tab Heat map of binding energy for S. miltiorrhiza compounds docking with core gene molecules, analyzed individually The lowest binding energy using hydrogen bonds was selected from the molecular docking results, which are visually represented in Fig. [80]4. As the energy becomes increasingly close to zero, the likelihood of activation increases; this is because the conformations of the ligand and the receptor are stable when they are in this state, which shows that the active component has some binding energy with the target if the binding energy (BE) of the active ingredient is -4.25 kcal/mol or less. In a broader sense, this suggests that the active ingredient has some binding energy. A good binding energy with the target is indicated by a binding energy (BE) of 5.00 kcal/mol or higher, whereas a strong binding energy is indicated by a BE of -7.00 kcal/mol or higher. By forming hydrogen bonds, including stabilizing and aromatic hydrocarbon hydrogen bonds, as well as π-π interactions, the active component S. miltiorrhiza has a significant binding affinity for the amino acid residues of the primary target. The bulk of the binding energies was approximately 5.0 kcal/mol, which validated the favourable docking outcomes. Fig. 4. [81]Fig. 4 [82]Open in a new tab Molecular docking pattern diagram of S. miltiorrhiza with some main targets against periodontitis Molecular dynamics results The baicalin-MMP9 and poriferasterol-TNF complexes were selected as typical pairings for molecular dynamics during 100 ns of simulation to further validate the stability of protein-ligand interactions. These parameters included the root mean square deviation (RMSD), the root mean square fluctuation (RMSF), the radius of gyration (Rg), the solvent-accessible surface area (SASA), and the hydrogen bonds (H-bonds)(Table [83]4and[84]5).As shown in Figs. [85]5A and [86]6A, RMSD analysis of the baicalin-MMP9 and poriferasterol-TNF complexes indicated that the protein–ligand systems reached equilibrium after 20 and 50 ns, suggesting stable binding between the bioactive compounds and their target atoms. Table 4. Associative free energy analysis (kcal/mol) VDWAALS EEL EPB ENPOLAR EDIPESAR ELTAG gas ELTAG solve Delta total Baicilin-MMP9 -40.99 -10.90 34.39 -3.21 0 -51.89 31.18 -20.71 Poriferasterol-TNF -49.07 -2.20 18.87 -4.65 0 -51.27 14.17 -37.09 [87]Open in a new tab Table 5. Average of RMSD, RMSF, RG, SASA and H hydrogen RMSD RMSF Rg SASA H-hydrogen ligand protein ligand protein Baicilin-MMP9 0.540725 0.57235 0.3561 0.8607 3.79655 6.36195 2–3 Poriferasterol-TNF 0.6147 0.9846 0.32825 0.977525 30.571575 86.588275 2 [88]Open in a new tab Fig. 5. [89]Fig. 5 [90]Open in a new tab MD of baicalin and MMP9, (A) Baicilin-MMP9-simulated RMSD changes; (B) Baicilin-MMP9-simulated RMSF changes; (C) Baicilin-MMP9 Rama Plot dynamic models; (D) Baicilin-MMP9 dynamic Rg models; (E) Baicilin-MMP9 hydrogen bond number dynamic models; (F) Baicilin-MMP9 2D free energy morphology; (G) Baicilin-MMP9 3D free energy morphology; (H) Residue energy of baicilin-MMP9 Fig. 6. [91]Fig. 6 [92]Open in a new tab MD of poriferasterol and TNF (A) Poriferasterol-TNF-simulated RMSD changes; (B) RMSF changes simulated by poriferasterol-TNF; (C) Rama Plot poriferasterol-TNF dynamic models; (D) Poriferasterol-TNF dynamic RG models; (E) Poriferasterol-TNF hydrogen bond number dynamic models; (F) Poriferasterol-TNF 2D free energy morphological distribution; (G) Poriferasterol-TNF 3D free energy morphological distribution; (H) Residue energy of Poriferasterol-TNF Throughout the simulation, RMSF was able to provide information regarding the degree of variability that each amino acid residue demonstrated. In the baicalin-MMP9 complex, residues 60–80 of MMP9 exhibited greater flexibility compared to other regions, suggesting localized variations in residue dynamics (Fig. [93]5B). In the poriferasterol-TNF complex, multiple regions of TNF exhibited residue flexibility, indicating dynamic structural changes during the simulation (Fig. [94]6B).SASA values for the baicalin-MMP9 complex were low throughout the simulation, indicating a stable binding interaction (Fig. [95]5C). In contrast, the SASA values for the poriferasterol-TNF complex were higher, suggesting a more flexible binding interaction (Fig. [96]6C).Rg values have been used to determine the degree of compactness. Values for the baicalin-MMP9 complex exhibited a decrease at 42 ns, followed by an increase at 50 ns; subsequently, they exhibited relative stability, indicating the overall structural integrity of the complex (Fig. [97]5D). In contrast, Rg values for the poriferasterol-TNF complex decreased at 5 ns and continued to decline, indicating relatively poor structural stability (Fig. [98]6D). Results of in vitro experiments S. miltiorrhiza’s effect on RAW264.7 cell viability RAW264.7 cells were subjected to different concentrations of S. miltiorrhiza (0, 0.01, 0.1, 1, 2, 3, 4, 5, and 9 mg/ml) to assess its impact on cellular activity. The findings demonstrated that lower concentrations (0.01 mg/ml, 0.1 mg/ml, and 1 mg/ml) markedly improved cell activity, but higher concentrations (2 mg/ml, 3 mg/ml, 4 mg/ml, and 5 mg/ml) exhibited no significant impact on cell activityP < 0.05). Treatment with the greatest quantity (9 mg/ml) suppressed cellular activityP < 0.05) (Fig. [99]7A). To further investigate the mechanism of S. miltiorrhiza in antiinflammatt periodontitis through cell experiments, we selected three concentrations of 0.01 mg/ml S. miltiorrhiza, 0.1 mg/ml S. miltiorrhiza and 1 mg/ml S. miltiorrhiza for subsequent experiments. Fig. 7. [100]Fig. 7 [101]Open in a new tab RAW264.7 inflammation-related factors were reduced by S. miltiorrhiza. (A)Effects of S. miltiorrhiza on RAW264.7 cell viability; (B) Impact of S. miltiorrhiza on RAW264.7 NO (C、D、E、F) Effects of S. miltiorrhiza on the levels of inflammatory factors (IL-1, 、IL-6, TNF-α, IL-10) in RAW264.7. Repeat three times per group (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001, suggesting a considerable difference from osteogenic induction medium-only cells S. miltiorrhiza prevented LPS-induced NO release in RAW264.7 cells NO content in the cell culture supernatant of S. miltiorrhiza treated samples was indirectly measured using the Griess method after 24 h of treatment. As illustrated in the figure, lipopolysaccharide (LPS), used as a positive control, markedly elevated NO levels. In contrast, S. miltiorrhiza treatment significantly suppressed NO production P < 0.05) (Fig. [102]7B). Impact of S. miltiorrhiza on inflammatory factor levels in RAW264.7 cells Based on quantitative real-time PCR results, pretreatment with S. miltiorrhiza at concentrations of 0.01 mg/mL, 0.1 mg/mL, and 1 mg/mL significantly reduced the mRNA expression levels of pro-inflammatory cytokines TNF-α, IL-6, and IL-1βP < 0.05). Conversely, the expression of the anti-inflammatory cytokine IL-10 was significantly increasedP < 0.05).The above results indicate that S. miltiorrhiza can not only inhibit the secretion of pro-inflammatory factors but also promote the expression of anti-inflammatory factors, suggesting that S. miltiorrhiza has potential anti-inflammatory properties(Fig. [103]7C and F). Discussion Periodontitis, a common chronic infectious condition affecting 50% of the global population, can severely compromise masticatory function, result in tooth loss, and induce facial collapse, considerably reducing quality of life [[104]14, [105]15]. The primary treatment for periodontitis focuses on eliminating and controlling inflammation in periodontal tissues to facilitate cell and tissue repair [[106]16]. One study concluded that drug therapy effectively treats periodontitis [[107]17]. Long-term drug treatment may result in drug resistance, bacterial imbalance, bone loss, and other adverse effects. S. miltiorrhiza, a traditional Chinese medicine that has been used for more than 2,000 years, contains bioactive components such as phenolic acids and tanshinones, which exhibit antioxidant and anti-inflammatory properties by inhibiting inflammatory mediators and enzymes [[108]18]. However, conclusive evidence for its efficacy in periodontitis remains lacking, prompting this study to employ network pharmacology, molecular docking, molecular dynamics, and cellular assays to explore its therapeutic potential. Network pharmacological analysis is extensively utilized to clarify the ways by which natural plant constituents address periodontitis and associated target investigations [[109]19]. This study identified 65 natural plant components (OB ≥ 30%, DL ≥ 0.18) and 132 active metabolite-related targets in S.miltiorrhiza. Cross-analysis identified 60 common targets between the selected natural plant components and periodontitis, suggesting potential activity against S. miltiorrhiza. The active ingredient-target-disease network indicated that Baicalin, Digallate, Luteolin, Poriferasterol, and Sugiol have higher degree values. Consequently, these key components were further analysed for their therapeutic effects on periodontitis. PPI network analysis revealed six potential key targets—AKT1, CASP3, IL-6, MMP9, STAT3, TNF, and TP53—that are significantly involved in the treatment of periodontitis with S.miltiorrhiza. Previous research has indicated that TNF-α is crucial for the inflammatory response in periodontitis, resorption of alveolar bone, and loss of connective tissue connection. TNF may cause periodontitis by weakening the oral mucosal barrier [[110]20]. In addition to its role in activating the NF-κB signalling pathway, the TNF molecule plays a significant role in facilitating the generation of inflammatory factors such as IL-6 and IL-11, and it also plays a role in exerting an influence on the inflammatory process that occurs within the body [[111]21, [112]22]. IL-6 acts as a proinflammatory agent in adaptive immunity and serves as an effective enhancer of inflammation [[113]23]. There is a high frequency of AKT1 activation in periodontitis, which serves to regulate inflammation and immunological responses [[114]24]. MMP9, which is primarily secreted by neutrophils and macrophages, is a proteolytic enzyme responsible for degrading collagen fibres and extracellular matrix components, thereby regulating tissue inflammation and disease. MMP9 primarily activates osteoclasts by removing collagen from demineralized bone, facilitating bone destruction. Consequently, its expression in periodontitis results in the destruction and reconstruction of periodontal tissue [[115]25–[116]27]. Additionally, computational simulations were used to explore the binding affinity and. mechanisms of core bioactive ingredients with key periodontitis-related targets. The molecular docking results revealed that the docking scores for the five components with the seven targets were all less than − 5 kcal/mol. Molecular dynamics simulations have demonstrated that the complexes formed between baicalin and MMP9, as well as between stigasterol and TNF, exhibit remarkable resistance to dissociation. Additional research, which included the examination of hydrogen bonds and the computation of binding free energy, has provided evidence that the interactions between these molecules are both feasible and powerful. Importantly, the simulations did not predict any key factors that would significantly violate the drug-like properties or lead to serious adverse and toxic reactions; this suggests a greater likelihood of success for these complexes in clinical settings. GO and KEGG enrichment analyses suggested that S. miltiorrhiza may target numerous signalling pathways for periodontitis therapy. These include the AGE-RAGE, TNF, PI3K-Akt, HIF-1, and IL-17 signalling pathways. The AGE-RAGE signalling pathway is activated by the inflammatory response, which promotes the release of inflammatory mediators, induces cell apoptosis, and leads to alveolar bone destruction [[117]28]. PI3K-Akt is a classic signalling pathway that regulates inflammation and cell differentiation. Studies have shown that bone morphogenetic protein-2 (BMP-2) can promote osteoblast differentiation by activating this pathway, providing a potential therapeutic strategy for periodontal tissue regeneration [[118]29]. Therefore, the effectiveness of S.miltiorrhiza is achieved through the regulation of various biological pathways. By evaluating the expression of inflammatory factors in RAW264.7 cells, S. miltiorrhiza was examined to determine whether it possessed the ability to prevent periodontitis.It has been demonstrated that S. miltiorrhiza effectively reduces the levels of TNF-α, IL-6, and IL-1β while upregulating the mRNA expression of the anti-inflammatory factor IL-10 in RAW264.7 cells. As a result, S. miltiorrhiza has the potential to regulate cellular inflammatory processes, which in turn may lessen the inflammatory response caused by LPS in RAW264.7 cells. S.miltiorrhiza’s ability to regulate periodontitis-related anti-inflammatory processes will be our main focus of future research, which will help us provide experimental and theoretical evidence as to how S.miltiorrhiza can be used to successfully treat and prevent periodontitis. This investigation should reveal further therapeutic potential of S. miltiorrhiza, leading to better periodontitis treatment strategies and a firmer theoretical base. Although this study provides valuable insights, several limitations should be acknowledged. First, the predictive results derived from network pharmacology and molecular docking require further validation through in vivo experiments. Second, the in vitro experiments focused exclusively on macrophages and did not assess other cell types critically involved in periodontitis pathogenesis, such as fibroblasts and epithelial cells. Future research should prioritize the following directions: (1) conduct animal studies to evaluate the therapeutic efficacy of S. miltiorrhiza in established in vivo periodontitis models; (2) investigate its antioxidant mechanisms through dedicated in vivo and in vitro experimental validation; and (3) explore its translational potential for treating other inflammatory diseases. Conclusion Within the context of the treatment of periodontitis, this study combines the investigation of S.miltiorrhiza with the emerging field of network science in order to investigate the active components of S.miltiorrhiza as well as the underlying molecular mechanisms that are involved. In light of the complexity of Chinese herbal medicine, particularly the multifaceted nature of its constituents and the therapeutic effects they produce, theoretical findings were further validated through the use of in vitro experiments. These experiments provide significant evidence of S.miltiorrhiza’s periodontitis therapy mechanism. Acknowledgements