Abstract Diabetic kidney disease (DKD) is a severe complication of diabetes, characterized by chronic inflammation and fibrosis. Tang Shen Ping Decoction (TSPD), a traditional Chinese medicine formulation, has shown therapeutic efficacy in DKD, yet its molecular mechanisms remain to be fully elucidated. To explore the multitarget mechanisms of TSPD, this study integrated network pharmacology, transcriptomic analysis, molecular docking, and molecular dynamics simulations, followed by in vivo and in vitro validation. A total of 248 active compounds and 649 potential targets of TSPD were identified, among which network pharmacology and transcriptomic integration highlighted 21 key genes involved in DKD pathogenesis. Protein–protein interaction network analysis further identified ALB, CCL2, EGF, FN1, and PTGS2 as central targets. Molecular docking confirmed strong binding affinities between core TSPD compounds, including quercetin and kaempferol, and these targets, particularly CCL2. Molecular dynamics simulations validated the stability of these interactions, identifying CCL2 as a crucial therapeutic target. In vivo experiments demonstrated that TSPD significantly improved renal function, attenuated fibrosis, and down-regulated CCL2, NF-κB, and TGF-β1 expression in DKD rats. In vitro, TSPD effectively suppressed CCL2/NF-κB activation and reduced the secretion of inflammatory cytokines (TNF-α, IL-6, and IL-1β) in high-glucose-treated HK-2 cells. Functional analysis confirmed that CCL2 overexpression exacerbated inflammation, while its silencing enhanced the anti-inflammatory effects of TSPD. These findings reveal that TSPD exerts renoprotective effects by targeting the CCL2/NF-κB axis, offering mechanistic insights into its anti-inflammatory and antifibrotic actions and providing a theoretical foundation for its clinical application in DKD treatment. __________________________________________________________________ graphic file with name ao5c01492_0013.jpg __________________________________________________________________ graphic file with name ao5c01492_0011.jpg 1. Introduction Diabetic kidney disease (DKD) is one of the most common and serious microvascular complications in patients with diabetes. According to a recent report by the International Diabetes Federation (IDF), approximately 463 million adults, or 9.0% of the world’s population, have diabetes, and 30–40% of these individuals are likely to develop DKD. DKD begins with increased microalbumin in the urine, progresses to proteinuria, and a gradual decline in kidney function, which can eventually lead to chronic kidney failure or even death. − Research has confirmed that inflammation plays a crucial role in the pathogenesis of DKD. In the diabetic state, factors such as hyperglycemia, altered renal hemodynamics, and impaired lipid metabolism stimulate the production of inflammatory mediators. These mediators trigger a series of inflammatory responses through pathways involving advanced glycation end products (AGEs) and protein kinase C (PKC), leading to pathological changes such as thickening of the glomerular basement membrane, extracellular matrix deposition in the glomerular mesangial area, and glomerulosclerosis. Therefore, DKD is considered to be an inflammatory disease. Given the critical role of inflammation in the pathogenesis of DKD, exploring traditional Chinese medicine strategies with anti-inflammatory effects is particularly important. Studies have shown that Traditional Chinese Medicine (TCM) can be effectively used in the treatment of DKD and has significant potential as a primary or adjunctive therapy. Tang Shen Ping Decoction (TSPD) consists of Pseudostellariae Radix (Taizishen, TZS), Astragalus membranaceus (Huangqi, HQ), Fructus corni (Shanzhuyu, SZY), Rehmanniae Radix Praeparata (Shudihuang, SDH), Dioscoreae Rhizoma (Shanyao, SY), Maydis Stigma (Yumixu, YMX), Smilacis Glabrae Rhizoma (Tufuling, TFL), Chuanxiong Rhizoma (Chuanxiong, CX), Salviae Miltiorrhizae Radix (Danshen, DS), Euonymi Alati Ramulus (Guijianyu, GJY), Rosae Laevigatae Fructus (Jinyingzi, JYZ), Hirudo medicinalis (Shuizhi, SZ), Bombyx Batryticatus (Jiangcan, JC), and Mantidis Ootheca (Sangpiaoxiao, SPX). Recent pharmacological studies have shown that HQ, SZY, SDY, SY, CX, DS, SZ, and their components exert therapeutic effects by inhibiting epithelial–mesenchymal transition, modulating inflammatory responses and oxidative stress, regulating glucose and lipid metabolism, and improving renal function. − In addition, TSPD has been shown to significantly reduce blood glucose, serum creatinine (Scr), blood urea nitrogen (BUN), and urinary albumin excretion rate (UAER) in patients with DKD, thereby alleviating disease symptoms. − Our previous research further confirmed that TSPD treatment significantly reduces renal function indicators such as BUN, Scr, and UAER, along with inflammatory cytokines TNF-α and IL-6 in DKD patients. However, the exact molecular mechanisms underlying the therapeutic effects of TSPD on DKD remain unclear. Further in-depth studies are needed to elucidate the mechanistic pathways by which TSPD exerts its nephroprotective effects. Network pharmacology is an emerging and popular research field that uncovers the complex mechanisms of TCMs by integrating bioinformatics, multiomics, and basic biological approaches. It employs multiple drug mechanisms that collectively target the same pathogenic signal transduction modules, thereby acting synergistically on key network proteins. This provides a novel avenue for investigating the multitarget, multicomponent effects of TCMs and has been extensively employed in the mechanistic studies of a multitude of diseases, including DKD. Network pharmacological analysis of TCMs for the treatment of DKD has identified a number of individual herbs and formulations that influence the progression of DKD, including Cordyceps sinensis, Salvia miltiorrhiza, Rhubarb, Shenyan Kangfu Tablets, PuRenDan, and Shenxiao Decoction. Nevertheless, to date, there have been no reports of network pharmacological research on TSPD for DKD. The objective of this study was to elucidate the intricate roles of the multifaceted components and targets of TSPD via network pharmacology. Furthermore, transcriptomics was employed to identify differential genes associated with DKD, with the findings integrated with network pharmacology to determine the core targets of TSPD linked to DKD with greater precision. Multiple enrichment analyses were subsequently conducted to explore the underlying molecular mechanisms, and the main compounds and targets were validated through molecular docking techniques. Finally, the specific mechanism by which TSPD improves fibrosis and inflammation was evaluated and confirmed in DKD rat and cell models. This study provides a theoretical basis for the clinical application of TSPD in DKD. 2. Materials and Methods 2.1. Screening and Target Prediction of Active Ingredients in TSPD The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, [41]https://tcmsp-e.com/) and the HERB database ([42]http://herb.ac.cn) were used to identify the chemical constituents of the 14 TCMs in TSPD. Among them, 11 TCMsTZS, HQ, SZY, SDH, SY, YMX, TFL, CX, DS, GJY, and JYZwere identified via the TCMSP database. The screening criteria applied in the TCMSP were oral bioavailability (OB) ≥30% and drug-likeness (DL) ≥0.18. Since JC, SPX, and SZ were not included in the TCMSP database, the HERB database was additionally used to retrieve and identify their chemical constituents. HERB integrates multiple traditional Chinese medicine databases and provides the most comprehensive list of herbs and their components to date. For these three ingredients, all the compounds related to JC, SPX, and SZ listed in the HERB database were included. This approach allowed us to identify the chemical constituents of all 14 ingredients in TSPD. The BATMAN-TCM database was subsequently used for target prediction, applying a score cutoff of 0.86 (LR = 112.67) and an adjusted P value of ≤0.05. The compound-related targets were obtained by integrating and deduplicating data from the TCMSP, HERB, and BATMAN-TCM databases. All obtained targets were standardized via the UniProt database ([43]https://www.uniprot.org/), with species restricted to Homo sapiens. Finally, the predicted targets were deduplicated and visualized in an ingredient–compound–target network via Cytoscape 3.10.2. The degree and radiality metrics were employed to identify the key active ingredients of TSPD. 2.2. Differential Expression Analysis of the [44]GSE96804 and [45]GSE99339 Data Sets The GEO data sets [46]GSE96804 and [47]GSE99339 were screened to select samples from healthy individuals and those with DKD. A total of 75 samples were included in the study, comprising 20 from healthy individuals and 55 from patients with DKD. The “sva” R package was employed for standardization and batch effect correction, and the “normalizeBetweenArrays” function was utilized for data normalization. Following these aforementioned normalization procedures, a comprehensive data set was constructed by extracting and combining the common genes. Differential expression analysis was conducted via the “limma” R package, with filtering conditions of |logFC| > 1 and an adjusted p value <0.05. The significant genes were then visualized via volcano plots and heatmaps. 2.3. Construction of Protein–Protein Interaction Network of Hub Genes Hub genes were identified on the basis of protein–protein interactions by intersecting the core target genes of TSPD with the differentially expressed genes (DEGs) from the transcriptome data set. The hub genes were imported into the STRING database ([48]https://string-db.org/), with the species set to “H. sapiens” for analysis. The resulting interaction networks were analyzed and visualized via Cytoscape 3.10.2, with degree values calculated. 2.4. Gene Function and Pathway Enrichment Analysis Gene Ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted via R packages such as “clusterProfiler”, “org.Hs.eg.db”, “Enrichplot”, “stringi”, “GOplot”, and “ggplot2”. The enrichGO function was employed for GO enrichment analysis, utilizing the org.Hs.eg.db database. Similarly, the enrichKEGG function was employed for KEGG enrichment analysis, with the KEGG database ([49]https://www.kegg.jp/) serving as the reference. A P value of less than 0.05 was employed as the screening criterion. If the q value (enrichment score) was less than 0.05, the corresponding functional terms and pathways were deemed statistically significant and retained. 2.5. GSEA and GSVA Analyses Gene set enrichment analysis (GSEA) is a comprehensive method for evaluating changes in gene sets by considering the enrichment of predefined sets across gene expression data. GSEA was conducted via data sets from the MSigDB database ([50]www.gsea-msigdb.org), the KEGG database, and the Reactome database ([51]https://reactome.org), with visualization facilitated by the OmicShare Analysis Platform ([52]http://www.omicshare.com/tools). Gene set variation analysis (GSVA) offers significant advantages in the context of individualized analysis, unsupervised data processing, and the capture of gene expression variations. In this study, the GSVA algorithm was employed to calculate a comprehensive score for each gene set, thereby enabling the assessment of potential biological function changes across different samples. For the “c2.cp.kegg_legacy.v2023.2.Hs.symbols.gmt” gene set, enrichment was deemed significant when the P value was less than 0.05. The GSVA algorithm was used to analyze data sets from healthy individuals and those with DKD via the R package “GSVA”. Pathways with a P value of less than 0.05 were identified as differentially expressed, and heatmaps were generated. 2.6. Molecular Docking Analysis The 2D structure files (Mol2 format) of the core active compounds were obtained from the TCMSP, and the 3D structure files (SDF format) of the core target proteins were retrieved from the Protein Data Bank (PDB) ([53]www.rcsb.org). The highest-resolution structure files were selected when possible. Molecular docking, dehydration, the removal of other heteroatoms, and automated docking visualization were performed via CB-Dock2. The binding affinity was evaluated via the AutoDock Vina score, which indicates the change in free energy associated with ligand–receptor binding. A negative value indicates a stable binding. The optimal docking conformation is indicated by the lowest AutoDock Vina score (kcal/mol). 2.7. Molecular Dynamics Simulation The receptor–ligand complexes obtained by molecular docking were further subjected to molecular dynamics (MD) simulations. Prior to the simulation, the complexes were preprocessed and separated using PyMOL. The General Amber Force Field (GAFF) was applied, and Sobtop was used to generate the initial configuration files (GRO files) for all small molecules, with ITP files defining the molecular force field parameters. The topological files for the proteins (ALB, CCL2, EGF, FN1, and PTGS2) were generated by using GROMACS (2020.3-MODIFIED). Prior to running the MD simulations, a comprehensive system preparation process was performed, including ionic, solvent, receptor, and ligand components. This preparation was performed in three main steps. First, energy minimization was performed using the steepest descent algorithm for 5000 steps to stabilize the system. Next, NVT (constant number of atoms, volume, and temperature) equilibration was performed by gradually heating the system to 300 K, while positional restraints were applied to the receptor and ligand. This phase lasted for 100 ps using the leapfrog integrator with a time step of 2 fs to maintain structural stability. In the third step, NPT (constant number of atoms, pressure, and temperature) equilibration was performed for 100 ps at a constant pressure of 1 bar and a temperature of 300 K to ensure system stabilization. After these preparatory steps, a 50 ns unconstrained MD production run was performed. Postsimulation analysis was performed using built-in GROMACS scripts to evaluate root-mean-square deviation (rmsd), root-mean-square fluctuation (RMSF), radius of gyration (R [g]), and hydrogen-bond interactions, providing insights into the dynamic stability and binding interactions of the receptor–ligand complexes. 2.8. Medicinal Preparation and Reagents TSPD was developed by Professor Yingming Qiu, a nationally recognized TCM expert at the Xiamen Hospital of Traditional Chinese Medicine. The TSPD preparation consisted of the following drug components and dosages: TZS (15 g), HQ (20 g), SZY (10 g), SDH (15 g), SY (15 g), SPX (10 g), YMX (30 g), TFL (30 g), CX (15 g), JC (10 g), DS (20 g), GJY (15 g), SZ (4 g), and JYZ (15 g). TSPD extracts were prepared according to a previously established method. All herbs were provided by Pientzehuang Honest (Xiamen, China). All of the herbs met the requirements of the Chinese Pharmacopoeia or the relevant herbal quality standards. The final concentration of raw TSPD at 100% yield was 1 g/mL. Irbesartan (Batch No. 2A386; 150 mg tablets; Sanofi) has been shown to delay the progression of DKD and was used as a positive control. Primary antibodies, including CCL2 (C–C motif chemokine ligand 2, Cat. No. ab283927), NF-κB (nuclear factor kappa-B, Cat. No. ab76302), TGF-β1 (transforming growth factor beta 1, Cat. No. ab21571), and GAPDH (glyceraldehyde-3-phosphate dehydrogenase, Cat. No. ab8245) used in animal experiments and secondary antibodies were purchased from Abcam. The primary antibodies Phospho-NF-κB p65 (cat. no. 3033) and NF-κB (Cat. No. 8242) used in cell experiments were purchased from Cell Signaling Technology, and CCL2 (catalog no. ab9669) was purchased from Abcam. Secondary antibodies were all purchased from Abcam. The ELISA kits were purchased from Beijing Baiaolaibo Technology Co., Ltd., including kits for the detection of rat tumor necrosis factor α (Cat. No. ZN2824), rat interleukin-6 (Cat. No. ZN2880), rat interleukin-1β (Cat. No. ZN2877), human tumor necrosis factor α (Cat. No. ZN2460), human interleukin-6 (Cat. No. ZN2272), and human interleukin-1β (Cat. No. ZN2236). UP (Urine Protein, Cat. No. C035-2-1), ALB (Albumin, Cat. No. A028-2-1), Cr (Creatinine, Cat. No. C011-2-1), BUN (Cat. No. C013-2-1), HbA1c (Glycated hemoglobin, Cat. No. H464-1-2), and INS (Insulin, Cat. No. H203-1-2) assay kits were purchased from the Nanjing Jiancheng Bioengineering Institute. 2.9. DKD Model Preparation and Experimental Grouping These experiments were conducted in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals (National Academies Press, Washington, DC, USA) and Animal Research: Reporting of In Vivo Experiments (ARRIVE) Guidelines and were approved by the Experimental Animal Ethics Committee of Fujian Medical University (Approval No. IACUC FJMU 2024-Y-1267). The experiment adhered to the principle of reducing the number of animals used and alleviating their discomfort. Sixty healthy male Wistar rats (8–10 weeks old, weighing 210–230 g) were housed in separate cages with an environmental temperature of 20 ± 2 °C, relative humidity of 60 ± 5%, noise lower than 85 dB, moderate light, and good ventilation. All rats were given 1 week of acclimatization with free access to food and water. A total of 50 rats were randomly selected for construction of the DKD model. To establish the model, 50 rats were randomly selected, and a diabetic kidney disease (DKD) model was constructed based on the method reported by Zhang et al., with slight modifications. Rats in the model group were fed a high-sugar and high-fat diet (HFD) for four consecutive weeks, consisting of 78.8% standard chow, 1% cholesterol, 0.2% bovine bile salt, 10% egg yolk powder, and 10% lard. At the beginning of week 4, a single intraperitoneal injection of streptozotocin (STZ, 30 mg/kg, dissolved in 0.01 mol/L citrate buffer, pH 4.5) was administered. Simultaneously, 5 mL of 50% glucose solution was given orally to prevent acute hypoglycemia. Rats in the control group received an equivalent volume of citrate buffer by oral gavage. After 72 h, blood glucose levels were measured by tail vein blood sampling. Rats with fasting blood glucose ≥16.7 mmol/L were considered to have successfully established diabetes. At the end of week 4, morning urine samples were collected to measure the urinary albumin-to-creatinine ratio (UACR). A UACR value of ≥30 mg/g was used as the diagnostic threshold for successful DKD model establishment. Control rats were maintained on standard chow throughout the experiment. In this study, all 50 rats initially enrolled for DKD model construction were successfully established based on the dual criteria of fasting blood glucose ≥16.7 mmol/L and UACR ≥30 mg/g. Subsequently, the rats were randomly assigned into six groups (n = 10 per group): normal control group (normal), model group (HFD/STZ), positive control group (Irbesartan, 25 mg/kg/day), and TSPD-treated groups at low, medium, and high doses. The dosages for each group of rats were calculated via a body surface area ratio conversion. The medium dose was calculated by converting the clinically recommended human daily dose of 224 g (crude herbs) for a 60 kg adult to an equivalent rat dose (based on 200 g of rat weight), yielding a medium dose of 23.06 g/kg/day. The low and high doses were defined as 50% and 200% of the medium dose, i.e., 11.53 and 46.12 g/kg/day, respectively. All doses were administered once daily by oral gavage for 12 weeks. The dosing period for all groups is 12 weeks. 2.10. Specimen Collection 24 h urine samples were collected at the end of the fourth and twelfth weeks of the experiment. At the end of the second, fourth, eighth, and twelfth weeks, blood glucose levels were quantified via blood collection from the tail vein in fasted rats for at least 12 h. At the end of the twelfth week, the rats were anesthetized intraperitoneally with 5% pentobarbital sodium (40 mg/kg). The abdominal cavity was then rapidly opened, and 2–3 mL of blood was extracted from the abdominal aorta. The serum was separated by centrifugation at 5000g for 10 min at room temperature. The samples were then placed at −80 °C for subsequent experiments. 2.11. Testing for Albumin, Creatinine, Urine Protein, Urea Nitrogen, Glycated Hemoglobin, and Insulin Levels The concentrations of urinary albumin (UAlb), urinary creatinine (UCr), urine protein (UP), ALB, serum creatinine (Scr), blood urea nitrogen (BUN), glycosylated hemoglobin (HbA1c), and insulin (INS) in the urine and serum of each group of rats were measured according to the instructions in the Nanjing Jiancheng Bioengineering Institute kit. 2.12. HE Staining The kidney tissues were fixed in 4% paraformaldehyde solution for 24 h, followed by gradient ethanol dehydration and clearing in xylene. The samples were then embedded in paraffin and sectioned to a thickness of 4 μm. After dewaxing and rehydration, the sections were stained with hematoxylin for 5–10 min, differentiated with ethanol hydrochloride, and subsequently stained with eosin for 1–3 min. The sections were then dehydrated in gradient ethanol, cleared in xylene, sealed, dried, and observed under a microscope. The glomerular area was quantitatively analyzed using ImageJ software based on HE-stained images to evaluate glomerular hypertrophy. 2.13. PAS Staining The procedure for PAS staining was similar to that for HE staining, with the following modifications: after fixation, dehydration, clearing, embedding, and sectioning, the sections were oxidized in 0.5% periodate solution for 5 min, stained with the Schiff reagent for 15 min, and then counterstained with hematoxylin for 1 min. The sections were subsequently dehydrated in gradient ethanol, cleared in xylene, mounted, dried, and observed under a microscope. The mesangial matrix expansion was quantified as a PAS-positive area fraction using ImageJ software. 2.14. Immunohistochemical Staining The kidney tissues were processed in the same manner as those used for the HE staining. The sections were then subjected to high-pressure antigen retrieval in citrate buffer for 10 min, treated with 3% hydrogen peroxide for 10 min, and blocked with 5% sheep serum for 30 min. The sections were incubated with primary antibody overnight, washed with phosphate-buffered saline (PBS), and incubated with a secondary antibody for 30 min. The color was developed via DAB, and hematoxylin was used for counterstaining. Gradient ethanol was used for dehydration, and xylene was used for clearing. The sections were then sealed, dried, and observed under a microscope. The positive area fractions of TGF-β1 and CCL2 were calculated using ImageJ software to evaluate the intensity and extent of the immunohistochemical staining. 2.15. Western Blot Analysis The total protein content of the kidney tissue and cell samples was extracted and quantified by the BCA method. Equal quantities of protein were separated by SDS-PAGE and transferred to a PVDF membrane. The membrane was then incubated with 5% skim milk powder for 1 h. The membranes were incubated with the primary antibody overnight, washed with TBST, and then incubated with the secondary antibody for 1 h. The color was developed and detected by imaging with an enhanced chemiluminescence (ECL) reagent. Band intensities were quantified using ImageJ software, and the relative expression levels of target proteins were normalized to GAPDH. Each experiment was performed in triplicate, and detailed original images are provided in [54]Figure S1A,B. 2.16. Cell Culture and Treatment The HK-2 cell line was purchased from ATCC. The medium was DMEM/F12 (Gibco, Cat. No. 11320033) supplemented with 10% FBS (Gibco, Cat. No. 11320033 and No. A5670701) and 1% penicillin/streptomycin (Gibco, Cat. No. 15140148). The cells were stimulated for 24 h in a high-glucose environment of 30 mmol/L to simulate the stress response induced by high glucose, with mannitol (Beyotime, Cat. No. ST2362) used as an osmotic control to prevent hyperosmolarity. To study the effects of CCL2 on HK-2 cells, Lipofectamine 3000 (Invitrogen, Cat. No. L3000015) dissolved in a serum-free medium was used for transfection. The cells were either silenced or overexpressed with si-CCL2 (MedChemExpress, cat. no. HY-RS02089) or pCMV-CCL2 (MiaoLing Biology, Cat. No. [55]P39235). TSPD was prepared at concentrations of 0.001, 0.01, 0.05, 0.1, and 1 g/mL, and the intervention lasted for 12 h. The results revealed that the optimal concentration was 0.1 g/mL. 2.17. RNA Extraction and Quantitative Real-Time PCR The total RNA was extracted from HK-2 cells via the TRIzol reagent (Invitrogen, Cat. No. 15596018), and the A260/A280 ratio was between 1.8 and 2.0. One microgram of total RNA was reverse transcribed into cDNA via a cDNA synthesis kit (Thermo Fisher Scientific, Cat. No. 4368814). qPCR was performed via SYBR Green qPCR Master Mix (Thermo Fisher Scientific, Cat. No. 4309155). The CCL2 sequences of primers used were as follows: forward primer (5′–3′): ACCAGCAGCAAGTGTCCCAAAG; reverse primer (5′–3′): TTTGCTTGTCCAGGTGGTCCATG. The GAPDH sequences of primers used were as follows: forward primer (5′–3′): ATGCCTCCTGCACCACCAACT; reverse primer (5′–3′): ATGGCATGGACTGTGGTCATGAGT. Data analysis was performed via the 2^–ΔΔCt method with GAPDH as the internal reference gene. Each sample was run in three biological replicates and three technical replicates. 2.18. ELISA Analysis The supernatant of the treated HK-2 cell culture was collected and centrifuged (1000g, 10 min) to remove the cell debris. The supernatant was stored at −80 °C until assayed. The manufacturer’s instructions were followed to assay and calculate the concentration of the target protein in the sample. 2.19. Statistical Analysis Bioinformatics analyses and data visualization were performed using R (version 4.3.2), while experimental data visualization was conducted with Prism (version 10.0, GraphPad Software). Statistical analyses were carried out using Prism, with results presented as mean ± standard deviation (SD). For comparisons between multiple groups, one-way analysis of variance (ANOVA) was performed to assess overall differences across the groups. Post hoc multiple comparisons were applied to identify specific group differences. A significance threshold of P < 0.05 was applied to determine statistical significance. 3. Results 3.1. Candidate Active Compounds in TSPD and Their Corresponding Targets A combination of the TCMSP and HERB databases was used to identify the compounds related to the 14 TCMs in the TSPD. A total of 1187 compounds were obtained in the TSPD, including 25 in TZS, 87 in HQ, 226 in SZY, 76 in SDH, and 71 in SY. Additionally, 29 compounds were found in SPX, 97 in YMX, 74 in TFL, 189 in CX, 4 in JC, 202 in DS, 24 in GJY, 35 in SZ, and 48 in JYZ. Since there were overlapping compounds among the 14 TCMs, a total of 248 unique active compounds were selected for further analysis after removing duplicates ([56]Figure A). Further details on the active chemical components of all of the herbs can be found in [57]Table S1. Additionally, 649 targets of TSPD were identified based on the target predictions from three databases, with duplicates removed for each herb, as shown in [58]Table S2. The ingredient–compound–target network was constructed via Cytoscape ([59]Figure B). Among the identified compounds, those with the most interactions across multiple targets were quercetin (MOL000098), luteolin (MOL000006), kaempferol (MOL000422), baicalin (MOL002776), and tanshinone IIA (MOL007154). These compounds were identified as the key active constituents of TSPD with potential therapeutic effects on DKD. 1. [60]1 [61]Open in a new tab TCMs, ingredients, and targets of TSPD. (A) Sankey diagram illustrating the relationship between the herbs and compounds of TSPD. (B) Network pharmacological analysis depicting the herb-compound-target network. The yellow nodes represent the 14 TCMs in TSPD, the orange nodes indicate the core compounds associated with TSPD, and the green nodes indicate the targets linked to TSPD. 3.2. Validation of Genes Associated with TSPD via the GeneCards and GEO DKD Data Sets To identify key genes related to TSPD and DKD, two transcriptome data sets, [62]GSE96804 and [63]GSE99339, were obtained from the GEO database to analyze differentially expressed genes (DEGs) between DKD patients and healthy individuals ([64]Table S3). First, the data sets were normalized to ensure comparability, and the results are shown in [65]Figure A,B. Differential gene analysis identified 188 DEGs, including 76 upregulated and 112 downregulated genes. These DEGs were then visualized via volcano plots ([66]Figure C) and heatmaps ([67]Figure D). To further refine the analysis, we identified 15,174 DKD-related targets by searching the GeneCards database. By intersecting the drug targets, DKD DEGs, and GeneCards DKD-related targets, 21 central genes were identified ([68]Figure E). A protein–protein interaction (PPI) network analysis of these 21 genes was then performed via the STRING database, and the results were visualized by degree via Cytoscape ([69]Figure F). This analysis identified ALB, CCL2, EGF, FN1, and PTGS2 as core targets associated with DKD. 2. [70]2 [71]Open in a new tab Differential gene expression, hub gene screening, and PPI analysis. (A,B) Box plots of gene expression normalization in control and DKD groups. (C) Volcano plot of DEGs (|log[2]FC| > 1, p < 0.05). (D) Heatmap of the top 40 DEGs, with colors indicating expression levels. (E) Venn diagram of TSPD targets (orange), DKD_GeneCards DEGs (green), and DKD DEGs (blue). (F) PPI network of 21 hub genes, with nodes representing genes and edges indicating interactions. 3.3. GO Biological Process and KEGG Pathway Analyses of Hub Genes To gain a deeper understanding of the molecular mechanisms involved in DKD, GO and KEGG enrichment analyses were performed on the hub genes ([72]Table S4). The results of the GO analysis, as illustrated in [73]Figure A, indicated significant enrichment in biological processes (BPs), cellular components (CCs), and molecular functions (MFs). The BP category was associated primarily with metabolic processes and stress responses, whereas the CC category was linked to the extracellular space, vesicles, and membrane-related structures. The MF category included activities such as fatty acid binding, antioxidant activity, and catalytic activity. KEGG analysis ([74]Figure B) revealed that the DEGs were enriched in several key metabolic and signaling pathways, including amino acid metabolism, drug metabolism, cell signaling, and inflammatory response. 3. [75]3 [76]Open in a new tab GO and KEGG analyses of hub genes. (A,B) Chord diagrams of GO (A) and KEGG (B) analyses, showing gene counts, enrichment, and pathway categories. (C,E) Bubble plots of GO analysis for the top 15 downregulated (C) and upregulated (E) hub genes. (D,F) Sankey diagrams of KEGG pathways for the top 15 downregulated (D) and upregulated (F) hub genes. For the downregulated genes ([77]Figure C,D), the GO analysis revealed that BPs were involved mainly in amino acid metabolism, cellular ketone metabolism, and oxidative detoxification. The CC category included structures such as peroxisomes and secretory granules, whereas the MF category included fatty acid binding, antioxidant activity, and vitamin B6 binding. KEGG pathway analysis revealed enrichment of pathways, such as amino acid metabolism, drug metabolism, the PPAR signaling pathway, cytochrome P450-mediated metabolism of exogenous substances, the IL-17 signaling pathway, and the TNF signaling pathway. The analysis of the upregulated genes ([78]Figure E,F) revealed that BPs were primarily involved in processes such as leukocyte chemotaxis, cytotaxis, and apoptotic cell clearance. The CC category focused on structures such as the collagen-containing extracellular matrix and lateral plasma membranes, whereas the MF category included chemokine activity and cell-to-cell adhesion mediator activity. KEGG analysis revealed enrichment of pathways, such as the NF-κB signaling pathway, the AGE–RAGE signaling pathway, the IL-17 signaling pathway, and the TNF signaling pathway. This study highlights the complex molecular processes underlying DKD, particularly the roles of metabolic regulation and inflammatory response. 3.4. GSEA and GSVA Analyses of Hub Genes To gain a comprehensive understanding of the molecular mechanisms of DKD, GSEA and GSVA were performed on whole transcriptome expression matrices. GSEA results, as shown in [79]Figure A–F, revealed a significant enrichment of several pathways and BP. Specifically, the GSEA-REACTOME analysis ([80]Figure A) revealed enrichment in lipoprotein assembly, mitochondrial translation, and metabolic transport. GSEA-KEGG analysis ([81]Figure B) revealed significant enrichment of the IL-1 pathway, MAPK pathway, ERK pathway, and JNK pathway in DKD. In addition, GSEA-GOBP, GOMF, and GOCC analyses ([82]Figure C–E) revealed significant alterations in BP, MFs, and CC, including amino acid metabolism, redox processes, and cellular membrane components. GSEA hallmark analysis ([83]Figure F) revealed a significant enrichment of pathways related to inflammatory response, immune regulation, and metabolic processes in DKD, and GSVA analysis ([84]Figure G) revealed distinct pathway activity patterns between DKD and control samples, further confirming the involvement of metabolic regulation and inflammatory signaling in the pathogenesis of DKD. Additional details are provided in [85]Table S5. These findings are consistent with the results of GO and KEGG analyses, reinforcing the critical role of metabolic dysregulation and inflammatory activation in DKD progression and the potential therapeutic targets. 4. [86]4 [87]Open in a new tab GSEA and GSVA analyses of all genes. (A–F) GSEA of REACTOME (A), KEGG (B), GOBP (C), GOCC (D), GOMF (E), and Hallmark (F) data sets, showing the top 10 enriched pathways (p < 0.05). (G) Heatmap of GSVA results for significantly enriched genes (p < 0.05). 3.5. Molecular Docking of Key Compounds and Targets To explore the molecular mechanisms by which TCM exerts therapeutic effects on DKD, molecular docking was conducted between five core targets (ALB, CCL2, EGF, FN1, and PTGS2) and five key TCM active ingredients (luteolin, quercetin, kaempferol, baicalin, and tanshinone IIA). The complete set of docking data is provided in [88]Table S6. The heatmap results revealed that all of the active ingredients exhibited stable binding interactions with their respective core targets, with Vina scores below −7 kcal/mol ([89]Figure A). Notably, luteolin had binding energies of −9.9 kcal/mol with ALB and −10.1 kcal/mol with CCL2. The highest binding energy was observed for quercetin with CCL2, with a value of −11.0 kcal/mol. Kaempferol exhibited a binding energy of −10.6 kcal/mol with CCL2, whereas baicalin had a binding energy of −11.0 kcal/mol with ALB. Tanshinone IIA had the highest binding energy of −12.2 kcal/mol with ALB. The specific molecular docking results are illustrated in [90]Figure , including those for ALB with tanshinone IIA ([91]Figure B), CCL2 with quercetin ([92]Figure C), EGF with luteolin ([93]Figure D), FN1 with baicalin ([94]Figure E), and PTGS2 with kaempferol ([95]Figure F). These results demonstrated that the core active components of TSPD have strong binding affinities with target proteins, validating the network pharmacology approach and suggesting that TSPD may ameliorate DKD by modulating the inflammatory response. 5. [96]5 [97]Open in a new tab Molecular docking of core compounds and targets. (A) Heatmap of docking results for five core compounds and five core targets, represented by the AutoDock Vina scores. (B) Molecular docking visualization of ALB with tanshinone IIA. (C) Visualization of the docking of CCL2 with quercetin. (D) Docking visualization of EGF with luteolin. (E) Docking visualization of FN1 with baicalin. (F) Docking visualization of PTGS2 with kaempferol. 3.6. Molecular Dynamics Simulation Analysis of Key Compounds and Targets Molecular dynamics simulations were conducted to further validate the binding stability of key compounds with the core targets. RMSD analysis ([98]Figure A) showed that ALB-Baicalin remained stable throughout 10–50 ns, while ALB-tanshinone IIA exhibited fluctuations near 40 ns. CCL2-quercetin remained stable across the full simulation, whereas CCL2-kaempferol was stable within 10–30 ns with minor fluctuations afterward. FN1-tanshinone IIA reached stability after 20 ns but showed slight variations. RMSF analysis ([99]Figure B) revealed that ALB and CCL2 maintained minimal structural fluctuations, whereas FN1 exhibited notable variations, indicating lower stability. Radius of gyration ([100]Figure C) indicated that ALB-baicalin and ALB-tanshinone IIA maintained stable compactness, while CCL2-quercetin and CCL2-kaempferol remained stable throughout, with FN1-tanshinone IIA showing minor fluctuations. Hydrogen-bond analysis ([101]Figure D) confirmed that ALB-baicalin and CCL2-quercetin maintained 1–5 hydrogen bonds throughout the simulation, while CCL2-kaempferol exhibited more dynamic hydrogen-bonding interactions. However, the EGF-tanshinone IIa and PTGS2-tanshinone IIa complexes exhibited poor stability, as detailed in [102]Figure S2A–D. 6. [103]6 [104]Open in a new tab Molecular dynamics simulation analysis of key compounds and targets. (A) RMSD plots of backbone atoms over simulation time for each key compound–target complex. (B) RMSF plots showing residue-level flexibility of the target proteins. (C) Radius of gyration (R [g]) plots depicting the overall compactness of the target–compound complexes along different axes. (D) Hydrogen-bond analysis displaying the number of hydrogen bonds formed between key compounds and targets during the simulation. The pink areas indicate the proportion of hydrogen bonds with a donor–acceptor distance ≤3.5 Å. 3.7. Comparison of the General Conditions of DKD Rats The rats in the experimental group exhibited a normal appetite, regular water intake, frequent activity, normal mental status, strong muscle tone, glossy fur, and normal urination and defecation. In contrast, the model group of rats presented symptoms such as emaciation, polyuria, polydipsia, polyphagia, reduced activity, depressed spirit, loose stools, and dull fur. In the treatment groups, including the irbesartan group and the low-, medium-, and high-dose traditional Chinese medicine groups, symptoms improved, although the mental state was still somewhat impaired, compared with that of the normal group. 3.8. Construction of the DKD Rat Model and the Effect of TSPD on Urinary Albumin and Creatinine Levels To evaluate the therapeutic effects of TSPD on DKD, a DKD rat model (HFD/STZ) was successfully established, as evidenced by significant changes in UAlb, UCr, and UACR compared to the normal group at both weeks 4 and week 12. UAlb levels were significantly elevated in the HFD/STZ group, indicating glomerular dysfunction. Treatment with irbesartan or TSPD at low, medium, and high doses significantly reduced UAlb levels in a dose-dependent manner, with substantial improvements observed by week 12 ([105]Figure A,B). UCr levels, which were reduced in the HFD/STZ group, reflecting renal tubular dysfunction, showed a partial recovery after treatment. By week 12, UCr levels in the TSPD and irbesartan groups approached those of the normal group, indicating improvement ([106]Figure C,D). Similarly, UACR, a critical indicator of renal function, was significantly elevated in the HFD/STZ group but was significantly reduced after TSPD treatment, with high-dose TSPD achieving effects comparable to those of irbesartan ([107]Figure E,F). These results demonstrate that TSPD ameliorates DKD-related renal dysfunction by improving UAlb, UCr, and UACR levels. 7. [108]7 [109]Open in a new tab Urinary levels of Alb, Cr, and UACR in different groups of rats. (A) UAlb levels at week 4. (B) UAlb levels at week 12. (C) UCr levels at week 4. (D) UCr levels at week 12. (E) UACR values at week 4. (F) UACR values at week 12. Statistical significance is indicated as follows: *p < 0.05, **p < 0.01, and ***p < 0.001 represent significant differences between the HFD/STZ and Normal groups. #p < 0.05, ##p < 0.01, and ###p < 0.001 indicate significant differences between all treatment groups and the HFD/STZ group. †p < 0.05 signifies significant differences between the medium-dose and high-dose groups compared to the low-dose group. 3.9. Effects of TSPD on the Renal Hypertrophy Index and Blood Functional Indices in the DKD Rat Model To evaluate the effects of TSPD on the renal function and metabolic status in DKD rats, the renal hypertrophy index, UP levels, and blood functional indices were measured. The results revealed a significant reduction in the renal hypertrophy index in the irbesartan group and the medium- and high-dose TSPD groups compared with the model group (P < 0.05), indicating alleviation of renal hypertrophy ([110]Figure A). The levels of UP, an indicator of renal damage, were significantly elevated in the DKD model group. Treatment with irbesartan and TSPD at all doses markedly reduced UP levels, particularly in the high-dose group (P < 0.05), suggesting improved renal function ([111]Figure B). Blood glucose levels were notably reduced in the irbesartan- and TSPD-treated groups, with the high-dose TSPD group showing the most significant improvement ([112]Figure C). Elevated INS levels in the DKD model group were significantly decreased by TSPD treatment, especially at medium and high doses (P < 0.05, [113]Figure D). Similarly, HbA1c levels decreased in the high-dose TSPD group, although this reduction was not statistically significant in the low- and medium-dose groups ([114]Figure E). Scr and BUN, key indicators of renal function, were significantly elevated in the DKD model group. Treatment with irbesartan and TSPD led to a significant reduction in the Scr level (P < 0.05), particularly in the high-dose group ([115]Figure F). Although BUN levels were not significantly reduced in the low- and medium-dose TSPD groups, a notable decrease was observed in the high-dose group (P < 0.05), indicating an improvement in the renal function ([116]Figure G). Therefore, TSPD, especially at higher doses, has a significant nephroprotective effect by reducing renal hypertrophy, lowering UP levels, and improving blood glucose and INS, Scr, and BUN levels. 8. [117]8 [118]Open in a new tab Renal hypertrophy indices, blood glucose changes, and blood biochemical indices in different groups of rats. (A) Histogram of the renal hypertrophy index among the different groups. (B) Histogram of UP among the different groups. (C) Line graph of blood glucose changes up to week 12 in different groups. (D) Histogram of insulin levels among the different groups. (E) Histogram of HbA1c among the groups. (F) Histogram of Scr among the different groups. (G) Histogram of BUN among the different groups. Statistical significance is indicated as follows: *p < 0.05 and **p < 0.01 represent significant differences between the HFD/STZ and Normal groups. #p < 0.05 and ##p < 0.01 indicate significant differences between all treatment groups and the HFD/STZ group. †p < 0.05 signifies significant differences between the medium-dose and high-dose groups compared to the low-dose group. 3.10. TSPD Exerts a Renoprotective Effect by Inhibiting Renal Fibrosis and the Inflammatory Response To explore the therapeutic effects of TSPD on renal fibrosis and inflammation, morphological changes in renal tissues were examined via HE and PAS staining. HE staining revealed intact glomeruli and renal tubules in the normal group, whereas the model group presented pronounced glomerular hypertrophy, an increased extracellular matrix, and mesangial cell proliferation. Treatment with irbesartan and various doses of TSPD significantly alleviated these pathological changes, with the high-dose TSPD group showing the most marked reduction in glomerular hypertrophy and extracellular matrix proliferation ([119]Figure A). PAS staining further revealed a marked increase in glycogen deposition, mesangial expansion, and basement membrane thickening in the model group. All of the treatment groups, with the exception of the low-dose TSPD group, improved these pathological changes, indicating that TSPD inhibits glycogen deposition and renal tubular dilation ([120]Figure A). 9. [121]9 [122]Open in a new tab Histopathological staining, immunohistochemistry, and Western blot analysis in different rat groups. (A) HE and PAS staining of the different groups (400× magnification). (B) IHC of TGF-β1 and CCL2 in different groups (400× magnification). (C) Quantification of the glomerular area (HE). (D) Quantification of the PAS-positive area fraction (PAS). (E) Quantification of the TGF-β1-positive area fraction (IHC). (F) Quantification of the CCL2-positive area fraction (IHC). (G) Western blot analysis of NF-κB, TGF-β1, and CCL2. (H) ELISA for TNF-α in the rat serum. (I) ELISA for IL-6 in the rat serum. (J) ELISA for IL-1β in the rat serum. Statistical significance is indicated as follows: *p < 0.05 and **p < 0.01 represent significant differences between the HFD/STZ and Normal groups. #p < 0.05 and ##p < 0.01 indicate significant differences between all treatment groups and the HFD/STZ group. †p < 0.05 signifies significant differences between the medium-dose and high-dose groups compared to the low-dose group. On the basis of network pharmacology, enrichment analysis, and a literature review, the CCL2 and NF-κB signaling pathways were selected to elucidate the mechanisms underlying the therapeutic effects of TSPD on DKD. Immunohistochemical analysis revealed a significant reduction in the expression of CCL2 and TGF-β1 in the renal tissues of the treatment groups compared with the model group, with the high-dose TSPD group exhibiting the most pronounced inhibitory effect ([123]Figure B). The quantitative results of HE, PAS, and IHC staining are shown in [124]Figure C–F. Western blot analysis further confirmed a substantial decrease in the protein expression of CCL2, NF-κB, and TGF-β1 in the treatment groups, with the high-dose TSPD group showing the most prominent effect ([125]Figure G). Additionally, ELISA revealed that TSPD treatment significantly reduced the levels of the inflammatory factors TNF-α, IL-6, and IL-1β in the rat serum, particularly in the high-dose group ([126]Figure H–J). These findings indicate that TSPD effectively inhibits the expression of CCL2 and NF-κB, reduces renal fibrosis, and decreases inflammation, thereby exerting a therapeutic effect on DKD. 3.11. TSPD Attenuates the Inflammatory Response via Modulation of the CCL2/NF-κB Signaling Pathway To investigate the molecular mechanism of TSPD in DKD, it was hypothesized that TSPD ameliorates DKD by regulating the CCL2/NF-κB signaling pathway, thereby alleviating the inflammatory response. HK-2 cells were subjected to high-glucose conditions, and CCL2 overexpression and knockdown were implemented, followed by TSPD intervention. The experimental groups were as follows: Control (Control), High Glucose (HG), High Glucose + CCL2 Overexpression (HG + oe-CCL2), High Glucose + CCL2 Silencing (HG+si-CCL2), High Glucose + TSPD (HG+TSPD), High Glucose + CCL2 Overexpression + TSPD (HG + oe-CCL2 + TSPD), and High Glucose + CCL2 Silencing + TSPD (HG + si-CCL2 + TSPD). First, the CCL2 mRNA expression was evaluated via qPCR ([127]Figure A,B). The results revealed a significant increase in CCL2 expression in the HG group compared with the control group, suggesting that high glucose induces CCL2 upregulation. TSPD treatment significantly suppressed this upregulation. In the HG + oe-CCL2 group, CCL2 mRNA levels were further increased; however, TSPD intervention (HG + oe-CCL2 + TSPD) effectively reversed this increase. In contrast, CCL2 mRNA expression was decreased in the HG + si-CCL2 group, and TSPD treatment further increased this decrease (HG + si-CCL2 + TSPD). These findings suggest that TSPD inhibits CCL2 expression in a high-glucose environment. 10. [128]10 [129]Open in a new tab TSPD modulates the CCL2/NF-κB signaling pathway and reduces inflammatory cytokine expression in different treatment groups. (A) Relative expression of CCL2 mRNA in the control, high glucose (HG), HG + TSPD, HG + oe-CCL2, and HG + oe-CCL2 + TSPD groups, as determined by quantitative PCR. (B) Relative expression of CCL2 mRNA in the control, HG, HG + TSPD, HG + si-CCL2, and HG + si-CCL2 + TSPD groups, as assessed by qPCR. The data are presented as the means ± SDs, with *p < 0.05 indicating statistically significant differences between groups. (C) Western blot analysis of p-NF-κB p65, NF-κB p65, and CCL2 protein levels across different treatment groups, with GAPDH used as a loading control. The adjacent bar graph presents the quantitative analysis of protein expression. The data are expressed as the means ± SDs. (D–F) ELISA results showing the concentrations of TNF-α (D), IL-6 (E), and IL-1β (F) in the supernatants of different treatment groups. The data are expressed as the means ± SDs. Statistical significance is indicated as follows: *p < 0.05, **p < 0.01, and ***p < 0.001 denote significant differences between the HG group and the control group. ^p < 0.05 and ^^p < 0.01 represent differences before and after TSPD treatment. #p < 0.05 and ##p < 0.01 indicate differences between HG + oe-CCL2 or HG + si-CCL2 groups and the HG group before and after treatment. †p < 0.05 denotes significant differences in the HG + oe-CCL2 and HG + si-CCL2 groups before versus after treatment. The activation of the NF-κB signaling pathway was subsequently assessed via Western blot analysis ([130]Figure C). Compared with the control group, the HG group presented significant activation of NF-κB phosphorylation and increased p-NF-κB p65 and CCL2 protein levels. TSPD treatment markedly reduced these protein levels. The overexpression of CCL2 in the HG + oe-CCL2 group led to further upregulation of p-NF-κB and CCL2, which was significantly attenuated by TSPD intervention (HG + oe-CCL2 + TSPD). Conversely, CCL2 silencing in the HG + si-CCL2 group substantially reduced p-NF-κB and CCL2 expression, and TSPD treatment (HG + si-CCL2 + TSPD) further potentiated this inhibitory effect. These results indicate that TSPD downregulates CCL2 expression and inhibits NF-κB activation, thereby modulating the CCL2/NF-κB signaling pathway. Finally, the levels of inflammatory cytokines in the cell supernatant were measured via ELISA ([131]Figure D–F). In the HG group, the levels of TNF-α, IL-6, and IL-1β were significantly increased, whereas TSPD treatment significantly reduced their expression. In the HG + oe-CCL2 group, the secretion of TNF-α, IL-6, and IL-1β was further increased but this increase was significantly mitigated by TSPD intervention (HG + oe-CCL2 + TSPD). Similarly, in the HG + si-CCL2 group, the levels of these cytokines were significantly decreased and TSPD treatment (HG + si-CCL2 + TSPD) further enhanced this inhibitory effect. These findings demonstrate that TSPD significantly reduces the level of secretion of inflammatory factors by modulating the CCL2/NF-κB signaling pathway. 4. Discussion DKD is a major complication of diabetes and a leading cause of end-stage renal disease (ESRD). Its progression can lead to a loss of kidney function, necessitating long-term dialysis or kidney transplantation. Improving therapeutic strategies for DKD is, therefore, crucial to prevent disease progression and alleviate patient burden. TCM has demonstrated unique efficacy in treating various diseases, and TSPD has shown significant clinical efficacy in managing DKD, particularly in its early stages. However, the precise therapeutic mechanisms of TSPD have been inadequately explored. This study integrated network pharmacology, transcriptomics, and molecular docking to elucidate the multitarget mechanisms of TSPD in treating DKD. Various enrichment analyses have indicated that TSPD can significantly ameliorate the pathological processes of DKD by modulating oxidative stress and inflammatory responses. In vivo and in vitro experiments further demonstrated that TSPD exerts its therapeutic effects by inhibiting the activation of the CCL2/NF-κB signaling pathway, thereby reducing inflammation and improving DKD pathology. These findings provide an initial understanding of the mechanisms underlying the efficacy of TSPD in DKD. TSPD therapy for DKD has been used in clinical practice for more than two decades. According to the TCM theory, the fundamental pathogenesis of DKD involves Qi and Yin deficiencies and internal stasis of dampness and turbidity. The treatment principle involves invigorating Qi, activating blood circulation, and dispelling dampness. TSPD is formulated on the basis of these principles, with the aim of simultaneously addressing the root cause and symptoms of the disease simultaneously. The formula includes Huangqi and Shudihuang to tonify the spleen and kidneys and Danshen and Chuanxiong to promote blood circulation and remove stasis. This approach aligns with the pathological characteristics of DKD, providing a theoretical foundation for the clinical application of the TSPD. Based on modern pharmacological investigations, this study further elucidated the potential therapeutic roles of the core active compounds in TSPD and their herbal sources in the treatment of DKD. In this study, 248 compounds in TSPD that interact with 649 targets were obtained through the TCMSP, HERB, and BATMAN-TCM databases, revealing the complex pharmacological network of TSPD. Core compounds such as quercetin, luteolin, kaempferol, baicalin, and tanshinone IIA exhibited significant interactions with multiple targets. These key compounds are primarily derived from the herbs GJY, HQ, JYZ, TFL, DS, TZS, and YMX included in the TSPD. Among them, HQ, a widely used Qi-tonifying herb in traditional Chinese medicine, has been reported to significantly improve albuminuria, proteinuria, and serum creatinine levels in DKD patients. Its bioactive constituents, such as quercetin, kaempferol, and baicalin, have demonstrated anti-inflammatory, antioxidant, and antifibrotic properties in multiple studies. In addition, preclinical studies have shown that GJY can ameliorate renal injury in DKD models by downregulating TGF-β1 expression. DS, which contains a high concentration of tanshinone IIA, has been shown to exert renoprotective effects by regulating the TGF-β/Smad and PI3K/Akt/FoxO signaling pathways, thereby inhibiting glomerulosclerosis and interstitial fibrosis. Furthermore, JYZ and TFL may act synergistically to support the therapeutic efficacy of TSPD. In addition, previous studies have also revealed the critical roles of the core active components of TSPD in the treatment of DKD. For instance, quercetin exerts nephroprotective effects by binding to PPARα to alleviate kidney injury. Luteolin reduces inflammation and oxidative stress by inhibiting the STAT3 pathway. Kaempferol mitigates apoptosis and promotes autophagy via the AMPK/mTOR pathway. Baicalin attenuates oxidative stress and inflammatory responses in DKD through the Nrf2 and MAPK pathways. Tanshinone IIA ameliorates diabetes-induced renal fibrosis by modulating the miR-34-5p/Notch1 axis. These findings highlight oxidative stress and inflammation as critical pathological mechanisms in chronic kidney disease (CKD), with these compounds exerting therapeutic effects through the modulation of these pathways. Molecular docking analysis further revealed that these compounds have binding energies below −7 kcal/mol with key targets, such as ALB, CCL2, EGF, FN1, and PTGS2, indicating stable interactions with these proteins. Evidence from previous studies, together with the findings of network pharmacology and molecular docking in this study, confirmed the stable binding interactions between the core active compounds of TSPD and key DKD-related targets, thereby providing further support for the therapeutic mechanism of TSPD through a multicomponent, multitarget synergistic regulatory network. To explore the mechanisms underlying the therapeutic effects of TSPD on DKD, the GeneCards database and transcriptomic differential gene analysis were utilized to identify 21 hub genes associated with TSPD and DKD. KEGG, GSEA, and GSVA pathway analyses revealed significant enrichment of these genes in the AGE–RAGE, NF-κB, IL-17, and TNF pathways, suggesting that these pathways play crucial roles in DKD pathogenesis. These signaling pathways have been extensively studied in the context of DKD pathophysiology and treatment. − It is hypothesized that TSPD exerts its therapeutic effects by modulating these pathways, thereby reducing inflammation and fibrosis. GO analysis further revealed significant enrichment of TSPD in metabolic processes, oxidative stress, and inflammatory responses, suggesting that TSPD may intervene in the key pathological processes of DKD through multitarget and multipathway regulation. Through molecular docking analyses and molecular dynamics simulations, CCL2 was identified as a key target of TSPD. CCL2 is shown to bind well to all of the core components by molecular docking. Notably, CCL2 maintained overall structural stability throughout the simulation, with minimal fluctuations upon ligand binding, supporting its role as a key and stable target in DKD. The strong and sustained interactions of CCL2-quercetin and CCL2-kaempferol, along with the preservation of CCL2’s structural integrity, further emphasize its therapeutic relevance in TSPD-mediated renal protection. These findings suggest that CCL2 plays a crucial role in DKD pathogenesis, and its targeted regulation via active compounds such as quercetin and kaempferol may underlie the anti-inflammatory effects of TSPD. Renal inflammation plays a crucial role in the onset and progression of DKD, and anti-inflammatory therapy holds promise for renoprotection in DKD. Studies have shown that CCL2 expression is significantly increased in DKD, leading to the accumulation of inflammatory cells in renal tissues and further aggravating kidney damage. Other studies have demonstrated that the downregulation of HMGN1 in diabetic kidneys attenuates tubular cell injury and protects against renal inflammation by suppressing CCL2 and KIM-1 expression through the TLR4 pathway. Moreover, enrichment analysis highlighted the importance of the NF-κB signaling pathway. Activation of the NF-κB and TGF-β1 pathways in DKD leads to increased expression of proinflammatory genes, including cytokines, chemokines, and adhesion molecules, promoting extracellular matrix accumulation in glomerular and tubular tissues and contributing to renal fibrosis. , Therefore, the focus was placed on the CCL2 and NF-κB signaling pathways to elucidate the molecular mechanisms by which TSPD ameliorates DKD. The DKD rat model experiments demonstrated that TSPD treatment reduced the levels of UAlb, UCr, UACR, UP, INS, and Scr, with significant effects observed particularly in the high-dose group. The HbA1c levels decreased significantly in the high-dose group, whereas no significant changes were observed in the other intervention groups, possibly due to HbA1c reflecting the average blood glucose level over a longer period. The 12 week treatment period may have been sufficient to exert effects only in the high-dose group. BUN levels can be influenced by factors such as renal function, diet, high fat intake, and dehydration, which explains the lack of significant differences in the other intervention groups except the high-dose group. HE and PAS staining revealed that TSPD ameliorated renal fibrosis in DKD rats. Western blot analysis revealed that TSPD inhibited the expression of CCL2 and NF-κB. The levels of inflammatory cytokines TNF-α, IL-6, and IL-1β also decreased significantly. These findings suggest that TSPD mitigates renal inflammation and fibrosis and improves renal function by inhibiting the CCL2/NF-κB pathway. The in vitro experiments further validated the mechanism of TSPD in treating DKD. Based on the preliminary findings from network pharmacology, transcriptomics, and molecular docking, it was hypothesized that TSPD may exert anti-inflammatory and renoprotective effects by modulating the CCL2/NF-κB signaling axis. In high-glucose-induced HK-2 cell models, CCL2 expression was significantly upregulated, accompanied by NF-κB activation, leading to elevated levels of inflammatory cytokines. TSPD intervention significantly inhibited the expression of CCL2 and the activation of NF-κB, reducing the secretion of inflammatory cytokines. Notably, in CCL2 overexpressing cells, although the inflammatory response was exacerbated, TSPD was able to partially reverse this effect, demonstrating its anti-inflammatory properties. Further, si-CCL2 intervention experiments showed that TSPD more effectively reduced NF-κB activity and inflammatory cytokine expression, indicating that CCL2 plays a critical role in the anti-inflammatory mechanism of TSPD. These results clearly demonstrate that TSPD exerts anti-inflammatory effects and ameliorates the inflammatory state in DKD by directly regulating the CCL2/NF-κB axis. In summary, this study revealed that TSPD can modulate the CCL2/NF-κB signaling pathway through multitarget and multipathway regulation, thereby inhibiting cellular inflammatory responses and improving DKD pathology. Despite the use of various modern scientific techniques to elucidate the potential mechanisms of TSPD in treating DKD, several limitations remain. First, the core components and targets of TSPD were screened from multiple databases, which may affect the generalizability and reliability of the results. Second, although some clinical research data on TSPD exist, the lack of large-scale clinical trial data limits a comprehensive evaluation of the clinical efficacy of TSPD. Furthermore, the detailed mechanisms of the core components of TSPD in treatment require further investigation. Future work will include analyzing the components of TSPD through experimental studies, utilizing chemical profiling techniques such as HPLC and MS, collecting clinical samples, and conducting metabolomic and proteomic research to fully elucidate the mechanisms of TSPD, providing robust scientific evidence for its application in modernized and precision medicine. 5. Conclusions This study systematically elucidated the multitarget mechanism of TSPD in the treatment of DKD by integrating network pharmacology, transcriptomics, molecular docking techniques, and molecular dynamics simulation. The core compounds of TSPDquercetin, luteolin, kaempferol, baicalin, and tanshinone IIAinteract with key targets such as ALB, CCL2, EGF, FN1, and PTGS2 to regulate oxidative stress, inflammatory response, and metabolic processes. In vitro and in vivo experiments further confirmed that TSPD exerts its therapeutic effect through the CCL2/NF-κB signaling pathway, effectively reducing the production of inflammatory factors and alleviating renal fibrosis. These findings provide a comprehensive understanding of the mechanism of TSPD in the treatment of DKD and offer a strong scientific basis for its clinical application. Supplementary Material [132]ao5c01492_si_001.pdf^ (1.6MB, pdf) [133]ao5c01492_si_002.xlsx^ (6.8MB, xlsx) Acknowledgments