Abstract Trifolin exhibits anti-tumor activities; however, its effect on hypertension remains unknown. This study was performed to investigate trifolin’s potential therapeutic effects and underlying mechanisms of action on angiotensin II (Ang II)-induced hypertension in mice and Ang II stimulated A7R5 cells. Mice were randomly allocated into six groups: control, Ang II, Ang II + Trifolin (0.1 mg/kg), Ang II + Trifolin (1 mg/kg), Ang II + Trifolin (10 mg/kg), and Ang II + Valsartan (10 mg/kg). The hypertensive mouse model was constructed by infusing Ang II via a micro-osmotic pump (500 ng/kg/min), and trifolin, valsartan, or double distilled water was administered intragastrically once daily for 4 weeks. Blood pressure, vascular function, pathological morphology, and collagen deposition in Ang II infused mice and cell viability of Ang II stimulated A7R5 cells were assessed. A networking pharmacology analysis was performed to identify potential targets, pathways, and processes. These were verified by determining proliferating cell nuclear antigen (PCNA) expression, cell migration, collagen protein expression and related pathway activation in vivo and in vitro using masson, immunohistochemistry, cell counting Kit-8 assays, phalloidin staining, wound healing assays, and western-blotting. Different concentrations of trifolin effectively mitigated the rise in systolic blood pressure, diastolic blood pressure, mean arterial pressure, pulse wave velocity, abdominal aorta wall thickness, and collagen deposition of Ang II infused mice. Notably, higher concentrations of trifolin exhibited greater attenuation which was similar to the effects of valsartan (a positive control). Networking pharmacology analysis identified 105 common targets and various gene ontology processes. The Kyoto Encyclopedia of Genes and Genomes pathways analysis identified multiple enriched signaling pathways, including responses to wounding, phosphatidylinositol 3-kinase complex, oxidoreductase, PI3K/AKT, and FoxO signaling pathways. Consistently, trifolin treatment significantly down-regulated the expression of PCNA and the ratio of p-PI3K/PI3K and p-AKT/AKT in the abdominal aorta tissues. In vitro study indicated that trifolin consistently reduced the cell viability, down-regulated the expression of PCNA, collagen I and collagen III, and reduced the cell migration, as well as reduced the ratio of p-PI3K/PI3K and p-AKT/AKT (similar with the effect of PI3K inhibitor: LY294002) in Ang II stimulated A7R5 cells. Trifolin treatment attenuated the elevation of blood pressure, the proliferation and collagen deposition of VSMCs, and modulated multiple signaling pathways, including PI3K/Akt pathway. These results suggest that trifolin could be a potential therapeutic approach for treating hypertension. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-01022-1. Keywords: Hypertension, Networking pharmacology, Trifolin, Vascular smooth muscle cells, PI3K/AKT Subject terms: Cardiovascular biology, Cardiology Introduction Hypertension is closely linked to cardiovascular disease and is considered a significant risk factor for global morbidity and mortality^[44]1. In a study involving 1,100,507 participants in low- and middle-income countries, 192,441 individuals were found to have hypertension. Of these, only a mere 10.3% had achieved control of their hypertension^[45]2. The prevalence of measures to control hypertension in China is relatively higher (13.0%) when compared to low-income countries, but the diagnosis, treatment, and control rates are still significantly lower than those of high-income countries (25%)^[46]3,[47]4. Therefore, identification of additional therapeutic approaches is urgently needed for hypertensive patients, particularly asymptomatic hypertensive patients, who are at an increased risk of the cardiovascular complications of damage to vital organs^[48]5. Due to a noticeable improvement in the effectiveness of treating hypertension when combined with western medicine, TCM has received growing interest^[49]6. Many formulas have been found to be effective in reducing blood pressure and alleviating hypertension-associated symptoms, as well as reducing hypertension-induced cardiovascular remodeling^[50]7,[51]8. Moreover, many natural compounds, such as quercetin and isoliensinine, have also been proven to have a good antihypertensive effect^[52]9–[53]12. Our previous studies have shown that Qingda Granule (QDG) play an antihypertensive role and reduce multiple target organs damage by regulating multiple signaling pathways^[54]13–[55]19. Further research revealed that many compounds in QDG exhibit antihypertensive effects, including alkaloids from nelumbinis plumula^[56]20, baicalin^[57]21, gastrodin^[58]22, neferine^[59]23, and Uncaria rhynchophylla^[60]24 and so on. However, the antihypertensive activity of many compounds is still unknown. Therefore, we thoroughly tested the antihypertensive activity of multiple compounds. Studies have shown that consuming flavonoids may lower the risk of cardiovascular-related deaths^[61]25,[62]26. Kaempferol 3-O-galactosyltransferase catalyzes the formation of trifolin by adding a galactoside molecule to kaempferol^[63]27. The flavonoid trifolin (kaempferol-3-O-galactoside) has been isolated from the aerial portions of Plumula nelumbinis^[64]28. Trifolin exhibits anticancer effects by inducing lung cancer cell apoptosis and inhibits tumor angiogenesis by modulating multiple pathways, including death receptor-dependent and mitochondria-dependent pathways^[65]29,[66]30. Moreover, upon network pharmacology analysis, trifolin was identified as a potential active component of QDG, even though its potential targets and pharmacological mechanisms of antihypertension remain unknown. To further explore the effects and underling mechanisms of trifolin on hypertension, we established both in vivo and in vitro models of Ang II-induced hypertension to investigate the impact of trifolin on blood pressure and vascular dysfunction, as well as pathological changes of the abdominal aorta. Network pharmacology was employed to predict the potential molecular targets, pathways, and processes involved in the treatment of hypertension by trifolin. These were further validated, both in vivo and in vitro, by determining pathway activation and related functional changes. Materials and methods Reagents Ang II and antibodies against PI3 kinase p85 alpha (phospho Y607; p-PI3K; catalog # ab182651) and PCNA (catalog # ab29) were obtained from Abcam (Cambridge, MA, USA). Eosin Y stain solution (catalog # G1100) and Cole’s hematoxylin solution (catalog no.# G1140) were provided by Beijing Solarbio Science & Technology Co., Ltd. (Beijing, China). Antigen repair solution (catalog # MVS-0066), Ultra-Sensitive™ SP (Mouse/Rabbit) immunohistochemistry (IHC) kit (catalog # KIT-9720), and 3,3’-Diaminobenzidine (DAB) substrate kit (catalog # DAB-0031) were provided by Maixin Biotechnology (Fuzhou, Fujian, China). Antibodies against PI3 kinase p85 (catalog # 4257), phospho-AKT (Ser473; catalog # 4060), AKT (catalog # 4691), and rabbit secondary antibody (catalog # 7074) were provided by Cell Signaling Technology (Danvers, MA, USA). Antibodies against p-PI3K (catalog # 41339) and p-AKT (catalog # 11054) for IHC staining were procured from Signalway Antibody (College Park, MD, USA). Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), trypsin-ethylenediaminetetraacetic acid, and bicinchoninic acid (BCA) protein assay reagent kits were provided by Thermo Fisher Scientific (Waltham, MA, USA). LY294002 was purchased from MedChemExpress (NJ, USA). Preparation of trifolin Trifolin (catalog # ABL2391), with a purity of ≥ 98%, was acquired from Chengdu Alfa Biotechnology Co., Ltd. (Chengdu, Sichuan, China). For the animal experiments, trifolin was dissolved in double distilled water (ddH2O) to the appropriate concentrations, and fresh solutions were prepared daily. For cell culture experiments, trifolin was dissolved in DMEM medium at a concentration of 100 mM. Animals and experimental protocols Male C57BL/6 mice aged 8 weeks were procured from Hangzhou Medical College (Certificate ID: SCXK 2019-0002; Hangzhou, China) and housed at the Animal Experiment Center of Fujian University of Traditional Chinese Medicine. The mice were kept in a pathogen-free environment under standard conditions at 24 °C ± 2 °C with a 12-hour light/dark cycle, and they were provided ad libitum access to food and water. To establish a hypertensive model, the mice were treated with Ang II via a micro-osmotic pump (500 ng/kg/min) and randomly allocated into six groups: control, Ang II, Ang II + Trifolin (0.1 mg/kg), Ang II + Trifolin (1 mg/kg), Ang II + Trifolin (10 mg/kg), and Ang II + Valsartan (10 mg/kg). The trifolin, dissolved in ddH2O, valsartan, dissolved in ddH2O, or plain ddH2O was infused intragastrically once daily for 4 weeks. Animal welfare and experimental protocols were performed in accordance with the ARRIVE guidelines and were applied in strict accordance with the guidelines of the Guide for the Care and Use of Laboratory Animals of the US National Institutes of Health. The animal experimental protocol for this study was approved by the Animal Care and Use Committee of Fujian University of Traditional Chinese Medicine (approval no. FJTCM IACUC 2022176) . Assessment of blood pressure Blood pressure was monitored using the CODA™ non-invasive blood pressure system (Kent Scientific; Torrington, CT, USA) by connecting a catheter to a pressure transducer. The systolic, diastolic, and MAP were measured while the mice were secured in a holder under dark and quiet conditions at 37 °C. Blood pressure was recorded prior to micro-osmotic pump implantation and on days 7, 14, 21, and 28 after implantation. Ultrasound imaging The Vevo 2100 high-resolution ultrasound instrument (FUJIFILM VisualSonics; Toronto, On, Canada) was used to conduct ultrasonic vascular assessments following the methodology described previously^[67]16. A 30 MHz probe was utilized to perform ultrasound scans while the mice were anesthetized with 1.5% isoflurane and positioned in a supine position on a heated table. B-mode and M-mode recordings were taken from below the xiphoid process, and measurements of the PWV and abdominal aorta thickness were measured and documented. H&E staining Abdominal aortic tissue was collected, immersed in a 4% paraformaldehyde solution for 48 h at room temperature, and then embedded in paraffin. Coronal sections with a thickness of 4 μm were prepared and subjected to H&E staining. Three images at 400× magnification were obtained from random locations of each tissue by using an intelligent automated optical microscope (Leica; Weztlar, Germany). Masson staining Paraffin sections were subjected to Masson trichrome staining. Representative images were captured using a light microscope (Leica) at a magnification of ×400 and collagen deposition was determined using Image J software. IHC staining After dewaxing, endogenous peroxidase activity was inhibited using an Ultra-Sensitive™ SP (Mouse/Rabbit) IHC kit, then washed with phosphate‑buffered saline. After blocking, the sections were incubated for 24 h at 4 °C with antibodies to PCNA (1:100), PI3K (1:400), p-PI3K (1:200), AKT (1:400) or p-AKT (1:200) overnight. These sections were then incubated with a biotinylated secondary rabbit/mouse antibody and horseradish peroxidase-labeled streptavidin. After the sections were examined by an intelligent automated optical microscope (Leica), protein expression was visualized using DAB and quantitatively analyzed by positive rate via Image J software (National Institutes of Health; Bethesda, MD, USA). Obtaining the targets for trifolin and hypertension Targets associated with hypertension were obtained via GeneCards ([68]https://www.genecards.org/). Additionally, DisGeNET ([69]https://disgenet.com/) was used to extract targets via the search term “hypertension.” Trifolin-related Targets were collected via PharmMapper ([70]https://www.lilab-ecust.cn/pharmmapper/) and Swiss-target prediction ([71]http://www.swisstargetprediction.ch/) databases. Constructing and analyzing PPIs PPI network data for common targets of trifolin and hypertension were retrieved using the STRING search tool ([72]https://cn.string-db.org/, Version 11.5). Cytoscape (Version 3.8.2) software was used to construct the “trifolin-hypertension-targets” interaction network. Exploration of functional annotations and pathways enrichment To determine the enriched GO functions and KEGG pathways, the DAVID tool was used to identify targets shared between trifolin and hypertension. Cell culture A7R5 cells were purchased from Procell Life Science & Technology Co., Ltd (Wuhan, Hubei, China; catalog # CL0316). All cells were maintained in DMEM with 10% FBS and 1% penicillin/streptomycin, kept in a humidified incubator at 37 °C with 5% CO[2]/95% air, and the medium was replaced every 48 h. Cell viability analysis A7R5 cells were suspended and added to a 96-well plate at a density of 0.3 × 10^5 cells/mL. Once the cells reached 40–50% confluence, they were deprived of serum for 6–8 h before being exposed to the different concentrations of trifolin for 24 h. Trifolin was administered at concentrations of 0, 25 µM, 50 µM, 100 µM, 200 µM, and 400 µM. Additionally, the A7R5 cells were treated with 1 µM Ang II and trifolin at concentrations of 0, 25 µM, 50 µM and 100 µM for 24 h. After the treatment period, 10 µL of Cell Counting Kit-8 was added to each well and incubated at 37 °C for an additional 2 h. Absorbance was measured at 450 nm using a microplate reader (Thermo Fisher Scientific; Waltham, MA, USA). Western blotting Total proteins were extracted from cells using western and immunoprecipitation cell lysis buffer. The cells were subjected to this buffer for 30 min on ice and then centrifuged at 12,000 g for 20 min at 4 °C. The supernatant, containing the extracted proteins, was collected, and protein concentration was measured using a BCA protein assay kit. An equal amount of protein was then subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis (10%) and transferred onto polyvinylidene difluoride (PVDF) membranes. To block any nonspecific binding, the PVDF membranes were incubated with blocking buffer at room temperature for 2 h. The membranes were then incubated with primary antibodies to p-PI3K, PI3K, p-AKT, AKT, PCNA, or GAPDH, all at a 1:1000 dilution, overnight at 4 °C. After washing with tris-buffered saline and tween 20 buffer to remove any unbound primary antibodies, the membranes were incubated with secondary rabbit/mouse antibody conjugated with horseradish peroxidase and diluted to a 1:5000 concentration. Protein bands were detected using a chemiluminescence kit, and the resulting data were analyzed using Image J software. Wound healing A7R5 cells were reseeded into 24-well plates (1 × 10^5 cells/well), and washed with PBS. A straight line was drawn through the cell monolayer using 10-µL pipette tip. Plates were incubated another 12 h, then photographed at magnification of 200× under a light microscope (Leica). Phalloidin staining A7R5 cells were treated and fixed with 4% paraformaldehyde, stained for 20 min with a conjugate of phalloidin and tetramethylrhodamine (AAT Bioquest, Sunnyvale, CA, USA), counterstained for 10 min with Hoechst solution, and photographed at a magnification of 200× under an UltraVIEW^® Vox fluorescence microscope (PerkinElmer, Santa Clara, CA, USA). Statistical analysis Statistical analyses were performed using SPSS 26.0 software (SPSS/PC+; Chicago, IL, USA). The Shapiro-Wilk test was utilized to determine normality. Mean ± standard deviation was used when the data were normally distributed, or median ± quartile was used to present the data. One-way analysis of variance was performed, followed by Bonferroni post hoc analysis, if the data were normally distributed and had equal variance. If the data were normally distributed but did not have equal variance, Games Howell post hoc analysis was used. The nonparametric Kruskal-Wallis test was applied when the data were not normally distributed. Repeated analysis of measurement variance was performed to analyze statistical significance of blood pressure. Statistical significance was set at p < 0.05. Results Trifolin attenuated blood pressure in Ang II-infused hypertensive mice A significant increase in systolic blood pressure (SBP; Fig. [73]1A), diastolic blood pressure (DBP; Fig. [74]1B), and mean arterial pressure (MAP; Fig. [75]1C) was observed in an Ang II-induced mouse model. However, administration of trifolin or valsartan effectively mitigated the rise in SBP (Fig. [76]1A), DBP (Fig. [77]1B), and MAP (Fig. [78]1C). The most substantial reduction in blood pressure was observed in the group that received 10 mg/kg of trifolin and valsartan. Additionally, there was no notable difference in weight among the six groups (Fig. [79]1D). Fig. 1. [80]Fig. 1 [81]Open in a new tab Impact of trifolin on blood pressure and body weight of angiotensin II (Ang II) infused mice. A tail-cuff plethysmograph method was used to measure blood pressure from each group: (A) systolic blood pressure, (B) diastolic blood pressure, (C) mean arterial pressure. (D) Body weight. Values are presented as mean ± standard deviation. Statistical significance is indicated as follows: ^ap < 0.05 vs. control group, ^bp < 0.05 vs. Ang II group, ^cp < 0.05 vs. Ang II + Trifolin (0.1 mg/kg) group, and ^dp < 0.05 vs. Ang II + Trifolin (1 mg/kg) group. N = 6 for each group. Trifolin alleviated vascular dysfunction and pathological changes in the abdominal aorta of Ang II-infused hypertensive mice Ultrasound was utilized to evaluate vascular function of the mice, and infusion of Ang II resulted in elevation of abdominal aorta pulse wave velocity (PWV), which was attenuated after trifolin and valsartan treatment (Fig. [82]2A–B). Furthermore, assessments using ultrasound (Fig. [83]2C) and hematoxylin and eosin (H&E) staining (Fig. [84]2D–E) demonstrated an increase in the thickness of the abdominal aortic wall of Ang II infused mice that was attenuated with the administration of trifolin and valsartan. Additionally, there was no notable difference in heart rate among the six groups (Supplementary Fig. [85]S1). Masson staining was revealed increased collagen content in Ang II infused mice that was attenuated with the administration of trifolin and valsartan (Fig. [86]2F-G).These above findings suggest that trifolin treatment could reduce Ang II-induced elavted blood pressure, vascular dysfunction and pathological changes. Fig. 2. [87]Fig. 2 [88]Open in a new tab Effects of trifolin on vascular function and pathological changes of the abdominal aorta in Ang II infused mice. Ultrasound and hematoxylin-eosin (H&E) staining was used to assess pulse wave velocity (PWV) and the thickness of the abdominal aortic wall in each group. (A) Representative ultrasound measurements of the abdominal aorta. (B) PWV in the abdominal aorta from each group as determined by ultrasound. (C) Abdominal aortic wall thickness as measured by ultrasound. (D) Abdominal aortic wall thickness as measured by H&E. (E) Representative abdominal aorta cross-sections as determined by H&E staining at a magnification of 400×. (F–G) Masson staining was performed to observe collagen deposition. The collagen content was calculated as the percentage of positive area relative to the total area. The scale bar is set at 50 μm. Values represented as mean ± SD. Statistical significance is indicated as follows: ^ap < 0.05 vs. control group, ^bp < 0.05 vs. Ang II group, and ^cp < 0.05 vs. Ang II + Trifolin (0.1 mg/kg) group. Analysis of protein-protein interaction (PPI) network highlights commonly shared targets between trifolin and hypertension In this study, a Circos diagram (Fig. [89]3A) was used to visualize the intersection of targets from multiple databases. Gene searches were executed using the terms “hypertension” and “trifolin” on the PharmMapper and Swiss databases. An overlapping gene assessment was conducted, and 105 common genes were identified (Fig. [90]3B). These 105 common targets were submitted to the Search Tool for Recurring Instances of Neighboring Genes (STRING) database to acquire data on target interactions. Network relationship data was obtained using a minimum required interaction score > 0.4 as a threshold. The resulting network consisted of 105 nodes and 1506 edges (Fig. [91]3C). To analyze the most potential genes, also known as hub genes, in the network, the Cytoscape integrated Cytohubba program was used. The top 15 hub genes were identified based on the Maximal Clique Centrality score, and a network with 15 nodes and 97 edges was constructed (Fig. [92]3D). These findings provide insight into the potential mechanisms underlying the effects of trifolin on antihypertension and may serve as a novel potential therapeutic approach for hypertension. Fig. 3. [93]Fig. 3 [94]Open in a new tab Networking pharmacology of common targets. (A) Circos plots displaying common targets between trifolin predicted targets and hypertension-related targets. Outer arcs represent the identity of each target, while inner arcs depict each target’s presence in various databases. Dark orange signifies targets found in multiple databases, while light orange represents those found in only one database. Purple lines connect identical targets found in specific databases, and blue lines connect targets with similar ontologies. (B) Depiction of common targets between trifolin predicted targets and hypertension-associated targets. (C) A network diagram presenting genes shared between trifolin predicted targets and hypertension-associated targets. (D) The top 15 hub genes with their corresponding Matthew’s Correlation Coefficient scores represented by a color gradient, with red representing high interaction and yellow representing lower interaction in the network. Clustering of common targets constructed network To gain insight into the structure and organization of complex networks, cauterization of the common network was performed, and five clusters were identified. Each cluster was characterized by a set of nodes, edges, and a score. The score is a measure of the strength of the clustering, with higher scores indicating more tightly connected nodes within the cluster. Cluster 1 consisted of 27 nodes and 372 edges, with a score of 14.308. This cluster contained the genes PLAU, APP, PIK3CA, and MMP9, which are involved in cell signaling, proliferation, and differentiation (Fig. [95]4A). Cluster 2 consisted of 15 nodes and 98 edges, with a score of 7.000. This cluster contained the genes PRKCD, EGFR, and TNF, which are involved in cell growth, differentiation, and apoptosis (Fig. [96]4B). Cluster 3 consisted of 5 nodes and 18 edges, with a score of 4.500. This cluster contained the genes COMT, ALDH2, and MAOA, which are involved in neurotransmitter metabolism and alcohol degradation (Fig. [97]4C). Cluster 4 consisted of 3 nodes and 6 edges, with a score of 3.000. This cluster contained the genes ADRA2A, PRKCB, and ADRA2C, which are involved in vasoconstriction, platelet aggregation, and protein kinase signaling (Fig. [98]4D). Cluster 5 consisted of 3 nodes and 6 edges, with a score of 3.000. This cluster contained the genes SHBG, GC, and F7, which are involved in hormone binding, vitamin D metabolism, and blood coagulation (Fig. [99]4E). Fig. 4. [100]Fig. 4 [101]Open in a new tab Cauterization of the network clusters highlighting common targets between proteins associated with hypertension and trifolin. The clusters are denoted as (A, B, C, D, and E), which stand for clusters 1, 2, 3, 4, and 5 respectively. The nodes with the highest level of interaction in clusters (A, B, and C) are highlighted in the center (KDR, PI3KR1, and MAOB) with middle and outer layer nodes. Assessment of GO and KEGG pathways Genetic ontology (GO) analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) database revealed that 105 common genes were enriched in numerous GO processes (false discovery rate cutoff 0.05). The top 30 processes of each GO category were displayed. These targets were enriched into multiple biological processes including the response to wounding and phosphorus metabolic regulation. Additionally, these were involved in the negative regulation of programmed cell death, positive regulation of intracellular signal transduction, cellular response to oxygen-containing compounds, and response to organonitrogen compounds (Fig. [102]5A). Further, the cellular processes with which the targets were associated included the phosphatidylinositol 3-kinase complex, endocytic vesicle lumen secretory granule lumen, transferring phosphorus-containing groups, and receptor complex (Fig. [103]5B). The molecular functions that these targets exhibited included insulin receptor substrate binding, calcium-dependent protein kinase C activity, oxidoreductase, ATP binding, and transmembrane signaling receptor activity (Fig. [104]5C). Moreover, these key target genes showed a close association with various Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including the vascular endothelial growth factor signaling pathway and the advanced glycation end-product (AGE)-receptor for AGE signaling pathway in diabetic complications, relaxin signaling pathway, FoxO, Rap1, and PI3K/AKT signaling pathways, and cancer and metabolic pathways (Fig. [105]5D). Fig. 5. [106]Fig. 5 [107]Open in a new tab Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis. GO function and KEGG pathway enrichment were performed based on the common targets. (A) Biological processes, (B) Cellular processes, (C) Molecular function, and (D) KEGG pathways. The y-axis depicts terms that are significantly enriched, while the enrichment score of these terms is shown on the x-axis. Trifolin attenuates up-regulated PCNA expression in vivo and in vitro, cell viability, and proliferation in Ang II-induced A7R5 cells Compared with the control group, the protein expression level of proliferating cell nuclear antigen (PCNA), a biomarker of proliferation, was significantly up-regulated in abdominal aortic tissue of Ang II infused mice; however, the increase was attenuated after trifolin and valsartan treatment (Fig. [108]6A). In vitro, CCK-8 analysis showed that trifolin did not affect cell viability at any of the concentrations tested (Supplementary Fig. [109]S2). Moreover, treatment with 25, 50, and 100 µM of trifolin significantly reduced the Ang II induced increase in cell viability (Fig. [110]6B–C). Similarly, Ang II stimulated expression of PCNA was reduced after trifolin treatment (Fig. [111]6D). Fig. 6. [112]Fig. 6 [113]Open in a new tab Effects of trifolin on the protein expression of proliferating cell nuclear antigens (PCNA) in vivo and in vitro, and proliferation of Ang II-stimulated A7R5 cells. (A) Immunohistochemistry (IHC) staining for protein expression of PCNA in the abdominal aorta of Ang II infused mice. Images were taken at a magnification of 400×. The scale bar is set at 50 μm. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, ^bp < 0.05 vs. Ang II group. N = 6 for each group. (B) Cell growth was observed using microscopy to determine cell confluence in Ang II stimulated A7R5 cells with or without trifolin treatment. Images were taken at a magnification of 200×. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, and ^bp < 0.05 vs. Ang II group. N = 3 for each group. (C) Cell viability was determined by Cell Counting Kit-8 assay, and the viability of untreated A7R5 cells was defined as 100%. (D–E) Protein expression of PCNA was examined by Western-blot, and protein expression was analyzed using Image J software. Glyceraldehyde 3-phosphate dehydrogenase expression was used as an internal control. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, and ^bp < 0.05 vs. Ang II group. N = 3 for each group. All values in this figure are presented as mean ± SD. Trifolin inhibits the formation of actin stress fibers and migration, attenuates up-regulated collagen I and collagen III in Ang II-induced A7R5 cells Treatment of cultured A7R5 cells with Ang II also induced the formation of actin stress fibers and migration (Fig. [114]7A–C), while also upregulating collagens I and III at the level of protein (Fig. [115]7D–F). However, trifolin partially reversed all these effects (Fig. [116]7A–F). These results suggest that trifolin reverses the fibrosis of actin cytoskeleton in aortic smooth muscle, the migration of smooth muscle cells, and the accumulation of collagen associated with hypertensive vascular dysfunction. Fig. 7. [117]Fig. 7 [118]Open in a new tab Effects of trifolin on actin stress fibers, migration and fibrosis in Ang II-induced A7R5 cells. The A7R5 cells were treated with 1 µM Ang II and trifolin at concentrations of 0, 25 µM, 50 µM and 100 µM for 24 h. (A) Phalloidin staining was used to detected the expression of actin stress fibers. A7R5 cells were stained with Hoechst (blue) and Phalloidin (red). Representative images were taken at a magnification of ×200. (B–C) Wound-healing assay was used to detect the migration of A7R5 cells. Representative images were taken at a magnification of ×200. n = 3 for each group. Protein expression of (D) collagen I and collagen III was examined by Western-blot. Protein expression was analyzed using Image J software (E–F). GAPDH expression was used as an internal control. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, and ^bp < 0.05 vs. Ang II group. N = 3 for each group. All values in this figure are presented as mean ± SD. Trifolin attenuated PI3K/AKT pathway activation in abdominal aortic tissue of Ang II infused mice The PI3K/AKT signaling pathway was explored to further explore the molecular underlying mechanism. Compared with the control group, the p-PI3K expression level, as well as the ratio of p-PI3K/PI3K, were significantly increased in the abdominal aortic tissue of Ang II infused mice; however, this increase was attenuated after trifolin and valsartan treatment (Fig. [119]8A–B). Moreover, trifolin treatment significantly attenuated increased expression of p-AKT and the p-AKT/AKT ratio in vivo (Fig. [120]8C–D). Fig. 8. [121]Fig. 8 [122]Open in a new tab Effects of trifolin on PI3K/AKT pathway activation in abdominal aortic tissue. Protein expression of (A) p-PI3K, (B) PI3K, (C) p-AKT and (D) AKT was examined by immunohistochemistry staining. Images were taken at a magnification of 200×. Protein expression is presented as mean ± SD. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, and ^bp < 0.05 vs. Ang II group. Trifolin attenuated PI3K/AKT pathway activation in Ang II-induced A7R5 cells PI3K/AKT pathway activation in vitro demonstrated that trifolin treatment significantly attenuated the increase of p-PI3K/PI3K and the p-AKT/AKT ratio in Ang II-stimulated A7R5 cells (Fig. [123]9A–D). Moreover, treatment of trifolin, LY294002 alone or combination of Trifolin and LY294002 significantly reduced the ratio of p-PI3K/PI3K and the p-AKT/AKT in Ang II stimulated A7R5 cells, while didn’t exhibits significantly difference among the above groups (Fig. [124]9E–H). The results confirmed that trifolin plays a protective role in hypertension dependent on the activation of PI3K/AKT signaling pathway. Fig. 9. [125]Fig. 9 [126]Open in a new tab Effects of trifolin on protein expression of the PI3K/AKT pathway in A7R5 cells stimulated by Ang II. The A7R5 cells were treated with 1 µM Ang II and trifolin at concentrations of 0, 25 µM, 50 µM and 100 µM for 24 h. (A–D) Protein expression of p-PI3K and PI3K and p-AKT and AKT was examined by Western-blot. Protein expression was analyzed using Image J software. GAPDH expression was used as an internal control. The A7R5 cells were treated with 1 µM Ang II and/or 100 µM trifolin, 25 µM LY294002 for 24 h. (E–H) Protein expression of p-PI3K and PI3K and p-AKT and AKT was examined by Western-blot. Protein expression was analyzed using Image J software. GAPDH expression was used as an internal control. Values in this figure are represented as mean ± SD. Statistical significance is indicated by the following symbols: ^ap < 0.05 vs. control group, and ^bp < 0.05 vs. Ang II group. Discussion The current status of the prevention and control of hypertension indicates the need to explore new therapeutic strategies. Therefore, it is necessary to further explore the role of TCM on cardiovascular disease. QDG is a well-known TCM formula designed by academician Keji Chen, which functions as an antihypertensive^[127]15–[128]18. We previously demonstrated that multiple components (including baicalin, gastrodin, neferine, uncaria rhynchophylla) of QDG have antihypertensive effects. However, the antihypertensive activity of many compounds is still unclear. Previously we performed a network pharmacology analysis, and trifolin was identified as a potential active component from QDG. However, its active potential targets and the pharmacological mechanism of its antihypertensive effects remain unknown. Hypertension is linked to structural and functional alterations in the vascular system and is a significant health concern^[129]31,[130]32. Trifolin, a flavone belonging to the flavonoid family, is commonly referred to as kaempferol-3-O-galactoside^[131]33, and it is found in multiple plants including Vicia faba^[132]33, Camptotheca acuminate^[133]34, and Consolida oliveriana^[134]35. It possesses antioxidative and anticancer properties, and it was found to reduce the levels of atherosclerosis and cardiovascular risk index^[135]30,[136]36,[137]37. To the best of our knowledge, this study is the first to investigate the therapeutic potential of trifolin for managing hypertension using an Ang II infusion mouse model, and it found that trifolin effectively attenuated the Ang II-induced increase in blood pressure, which indicates its potential as an antihypertensive agent. Since vascular tone plays a crucial role in regulating blood pressure and blood flow^[138]38, we further evaluated functional changes of the abdominal aorta in vivo using ultrasound, and demonstrated that trifolin treatment significantly reduced Ang II-induced PWV and attenuated pathological changes in the abdominal aortic vessel wall. Overall, our findings suggest that trifolin treatment can alleviate both functional and pathological changes in the abdominal aorta of Ang II-infused mice, providing further evidence for its therapeutic potential in the treatment of hypertension. The current study further confirmed that trifolin treatment attenuated Ang II-induced the elevation of blood pressure, which was similar to the positive control valsartan (a kind of ARBs using as a positive control), suggesting the potential of trifolin to serve as an anti-hypertension medicine. However, the anti-hypertension effect of trifolin should be further verified in different animal models, including two-kidneys-one-clip and deoxycorticosterone acetate, and its underlying mechanisms should be further explored in depth. Network pharmacology is a novel field that builds upon the principles of systems biology. This involves the analysis of networks within biological systems and the identification of specific signaling nodes that can be targeted to develop a multi-target drug molecule network^[139]39. To comprehensively investigate the potential mechanism by which trifolin attenuates blood pressure elevation using network pharmacology analysis, this study identified 1036 trifolin-related target genes, 1947 hypertension-related genes, as well as 105 related common genes. These genes were significantly enriched in several GO processes, including intracellular signal transduction, programmed cell death, and others. Moreover, analysis of the topological variables of the network revealed that genes from the PI3K and protein serine kinase genes were highly ranked throughout the network, especially PIK3CA. PIK3CA encodes the p110α catalytic subunit of PI3K which through its role in the PI3K/AKT pathway is important for the regulation of important cellular functions such as proliferation, metabolism and protein synthesis, angiogenesis and apoptosis^[140]40. On the other hand, PIK3Rl, which encodes the regulatory subunit alpha of PI3Kmodulates PI3K’s catalytic activity and facilitates downstream activation of the AKT signaling pathway^[141]41,[142]42. This pathway is crucial for vascular smooth survival muscle cell proliferation, migration, and dysregulated expression or activity of PIK3Rl has been implicated in vascular pathologies, including endothelial dysfunction, oxidative stress, and vascular fibrosis^[143]43,[144]44. In addition, KEGG analysis analysis indicated that vascular endothelial growth factor signaling pathway, FoxO, Rap1, and PI3K/AKT signaling pathways were enriched, which have been widely reported to play vital functional roles in hypertensive vascular diseases^[145]17,[146]45–[147]47. Based on the target genes and potential pathway analysis that trifolin may exert its beneficial effects on blood pressure regulation through multiple signaling pathways, notably including the PI3K/AKT signaling pathway. Vascular smooth muscle cells (VSMC) proliferation, migration and fibrosis that results in vascular remodeling is a hallmark of vascular pathology including hypertension. To validate the network pharmacology results, we conducted both in vivo and in vitro experiments, which confirmed the antihypertensive effect of trifolin. Activated PI3K helps guide AKT to the plasma membrane where it is phosphorylated, and this contributes to the development of hypertension^[148]48. Therefore, targeting PI3K/AKT pathway activation significantly inhibits the proliferation and phenotypic conversion of VSMC, thereby inhibiting vascular remodeling and controlling blood pressure changes, which suggests a promising potential therapeutic strategy for managing hypertension^[149]20,[150]49–[151]53. In our current study demonstrated that trifolin consistently reduced Ang II stimulated A7R5 cell viability, down-regulated PCNA, collagen I and collagen III, reduced the cell migration and phenotypic transformation. However, we will further explore the effect of trifolin in adhesion, immunity, connection and diastole function of endothelial cells induced by hypertension. This study demonstrated that trifolin inhibited the proliferation and collagen deposition of Ang II-induced A7R5 cells and significantly decreased the expression of p-PI3K and p-AKT in Ang II-stimulated A7R5 cells and the abdominal aorta of Ang II infused mice. These results suggest that the antihypertensive properties of trifolin may be attributed to the inhibition of the PI3K/AKT pathway activation. These results justify further investigation into this traditional Chinese medicine compound as a treatment against hypertension, and they identify several molecules and pathways worth exploring in order to optimize this medicine and develop other anti-hypertensive drugs. Our results suggest that trifolin is a potential agent for treating hypertension patients and a promising drug for PI3K/AKT research. Based on network pharmacology analysis, more work is needed to further explore the underlying mechanisms of trifolin in sequenced potential target genes and other signaling pathways including redox enzymes, the FoxO and ERK signaling pathway, which indicated that the regulation and mechanism of vascular dysfunction. In addition, primary vascular smooth muscle cells in C57BL/6 mice of abdominal aorta should be used to investigate the effect of the treatment of trifolin on phenotypic switch (α-SMA, OPN), fibrosis (mmp2, mmp9) of smooth muscle cells after Ang II stimulation in future study. Our results should be verified and extended in other animal models of hypertension, and the mechanism of action of trifolin should be elucidated in detail to allow its optimization as an anti-hypertensive compound and inspire the design of similar compounds. Conclusion Trifolin treatment attenuated the elevation of blood pressure, the proliferation and collagen deposition of VSMCs, and modulated multiple signaling pathways, including PI3K/Akt pathway. These results provide experimental evidence for the therapeutic potential of antihypertensive effects of trifolin. Electronic supplementary material Below is the link to the electronic supplementary material. [152]Supplementary Material 1^ (115.7KB, docx) [153]Supplementary Material 2^ (1.8MB, pdf) Abbreviations Ang II Angiotensin II BCA Bicinchoninic acid BSA Bovine serum albumin CVDs Cardiovascular diseases CCK-8 Cell Counting Kit-8 DBP Diastolic blood pressure DAVID Database for annotation visualization and integrated discovery DAB Diaminobenzidine ddH[2]O Double distilled water DMEM Dulbecco’s modified eagle medium FBS Fetal bovine serum GO Gene ontology GAPDH Glyceraldehyde 3-phosphate dehydrogenase H&E Hematoxylin and eosin IF Immunofluorescence KEGG Kyoto encyclopedia of genes and genomes MAP Mean arterial pressure PBS Phosphate-buffered saline PVDF Polyvinylidene fluoride membrane PWV Pulse wave velocity PPI Protein-protein interaction QDG Qingda granule STRING Search tool for recurring instances of neighboring genes SBP Systolic blood pressure TSV Tab-separated value TCM Traditional Chinese medicine VSMCs Vascular smooth muscle cells Author contributions All authors contributed to the study conception and design. Jun Peng, Min Yu, and Keji Chen conceived and designed the experiments. Aling Shen, Meizhu Wu, Zhi Guo, and Jinkong Wu conducted the animal experiments and analyses. Farman Ali and Aling Shen performed network pharmacology analysis. Meizhu Wu, Zhi Guo, and Ying Cheng conducted H&E and IHC staining. Aling Shen, Hong Chen, and Meizhu Wu conducted the cell experiments and Western blotting. Aling Shen, Dawei Lian, and Meizhu Wu analyzed the data. Aling Shen, Meizhu Wu, and Farman Ali drafted the manuscript, and all authors commented on preliminary versions of the manuscript. Jun Peng, Min Yu, and Keji Chen provided the materials and revised the manuscript. All authors read and approved the final manuscript. Funding This study was sponsored by the National Natural Science Foundation of China (U22A20372 and 82074363), Fujian Research and Training Grants for Young and Middle-aged Leaders in Healthcare, the Science and Technology Major Project of Fujian Province (2019YZ014004), the Development Fund of Chen Keji Integrative Medicine (CKJ2020003), the Young Elite Scientists Sponsorship Program of the China Association of Chinese Medicine (2021-QNRC2-B19), and the Youth Talent Support Program from Fujian University of Traditional Chinese Medicine (XQB202202). Data availability The datasets used and/or analysed during the current study available from the corresponding author on reasonable request. Declarations Competing interests The authors declare no competing interests. Ethical approval Animal welfare and experimental protocols were applied in strict accordance with the guidelines of the Animal Care and Use Committee of Fujian University of Traditional Chinese Medicine (Approval No. FJTCM IACUC 2022176). Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Aling Shen and Meizhu Wu these have contributed equally to this work and share first authorship. Contributor Information Jun Peng, Email: pjunlab@hotmail.com. Min Yu, Email: yumin@sinopharm.com. Keji Chen, Email: Kjchenvip@163.com. References