Skip to main content
Pharmaceutical Biology logoLink to Pharmaceutical Biology
. 2023 Feb 24;61(1):473–487. doi: 10.1080/13880209.2023.2168020

PI3K/AKT/SERBP-1 pathway regulates Alisma orientalis beverage treatment of atherosclerosis in APOE−/− high-fat diet mice

Ruiyi Liu a,b,c,*, Yan Sun a,b,*, Dong Di a,b, Xiyuan Zhang d, Boran Zhu a,b,, Haoxin Wu a,b,
PMCID: PMC9970249  PMID: 36825364

Abstract

Context

Previously, we found Alisma orientalis beverage (AOB), a classic traditional Chinese medicine (TCM) formulation, had the potential effect of treating atherosclerosis (AS). The underlying mechanism was still unclear.

Objective

As an extention of our previous work, to investigate the underlying mechanism of action of AOB in the treatment for AS.

Materials and methods

Network pharmacology was conducted using SwissTargetPrediction, GeneCards, DrugBank, Metascape, etc., to construct component-target-pathway networks. In vivo, AS models were induced by a high-fat diet (HFD) for 8 consecutive weeks in APOE−/− mice. After the administration of AOB (3.8 g/kg, i.g.) for 8 weeks, we assessed the aortic plaque, four indicators of blood lipids, and expression of the PI3K/AKT/SREBP-1 pathway in liver.

Results

Network pharmacology showed that PI3K/AKT/SREBP-1 played a role in AOB’s treatment for AS (PI3K: degree = 18; AKT: degree = 17). Moreover, we found that the arterial plaque area and four indicators of blood lipids were all significantly reversed by AOB treatment in APOE−/− mice fed with HFD (plaque area reduced by about 37.75%). In addition, phosphorylated expression of PI3K/AKT and expression of SREBP-1 were obviously increased in APOE−/− mice fed with HFD, which were all improved by AOB (PI3K: 51.6%; AKT: 23.6%; SREBP-1: 40.0%).

Conclusions

AOB had therapeutic effects for AS by improving blood lipids and inhibition of the PI3K/AKT/SERBP-1 pathway in the liver. This study provides new ideas for the treatment of AS, as well as new evidence for the clinical application of AOB.

Keywords: Traditional Chinese medicine, high-fat diet, aortic plaque, lipid metabolism, network pharmacology

Introduction

The latest data from the Global Burden of Disease (GBD) showed that approximately 18.6 million people worldwide died of cardiovascular disease in 2019, which has surpassed infectious diseases as the leading cause of death and disability worldwide (Roth et al. 2020). Atherosclerosis (AS) is the main factor leading to the global epidemic of cardiovascular and cerebrovascular diseases, which is mainly characterized by lipid deposition and chronic inflammation in the arterial wall (Roth et al. 2020). Atherosclerosis (AS) is characterized by fibrofatty lesions formed on the inner wall of arteries and is the primary pathological basis of cardiovascular and cerebrovascular diseases (Kobiyama and Ley 2018). Increasing evidence has indicated that hypercholesterolemia-induced vascular inflammation and cholesterol deposition together constitute a risk factor for AS (Koeth et al. 2019). Statins, lipid-lowering drugs such as atorvastatin and rosuvastatin, are the first-line drugs for the treatment of AS in modern medicine and they have significant clinical efficacy in lowering blood lipids, but they can also cause adverse reactions such as liver damage and rhabdomyolysis (Soppert et al. 2020; Aryal et al. 2021). The latest study showed that a serine protease, PCSK9, actively targets LDL-R and causes its excessive accumulation, while PCKS9 inhibitors significantly reduce LDL-C levels and reverse plaque-like changes (Solanki et al. 2018). However, the high cost of the compound and lack of long-term safety and efficacy data limit its use in patients. Therefore, there is an urgent need to find drugs with safe effects and better efficacy.

Hypercholesterolemia is recognized as the main factor leading to AS; reducing blood cholesterol levels is an important way to prevent the development of AS (Francis 2010). Various studies suggested that lipid metabolism mechanisms play a key role in the pathophysiology of AS and that elevated LDL cholesterol leads to AS independent of inflammation, whereas residual cholesterol can drive the inflammatory component of AS (Geovanini and Libby 2018). However, this evidence was not capable of solving the root cause of treating AS. Alisma orientalis beverage (AOB) was first recorded in ‘Huangdi Neijing’, an ancient Chinese medical book, consisted of three herbs including Alismatis rhizoma (Sam.) Juzep. (Alismataceae) (Zexie), Atractylodis macrocephalae rhizoma Koidz. (Asteraceae) (Baizhu), and Pyrolae calliantha H. Andres (Pyrolaceae) (Luxiancao) based on the Chinese Pharmacopeia (2020 Edition). A previous study found that AOB can effectively inhibit the progression of atherosclerosis and improvement of blood lipid levels, and its mechanism of mitigating atherosclerosis may be related to gut microbiota and its metabolite (Zhu, Zhai, et al. 2020). However, how AOB influenced blood lipid levels was not identified. Due to the complex components of this formula, it is difficult to explore multiple targets in traditional Chinese medicine (TCM) formulation. Therefore, the underlying mechanism of AOB’s therapeutic actions have not been fully elucidated.

TCM has characteristics of multi-component, multi-target and integrity. Network pharmacology is based on theories of systems biology, genomics, proteomics and other disciplines, using high-throughput omics data analysis, computer simulation and network database retrieval (Hopkins 2008). The technology reveals the network relationship of drug-gene-target-disease interactions, predicts the mechanism of action of drugs through network relationships, evaluates drug efficacy, adverse reactions, etc., explores essential attributes of TCM by referring to the research ideas of network pharmacology, and has achieved good preliminary results in revealing the comprehensive overall effect of multiple pathways, multiple targets and multiple components of TCM (Zeng et al. 2019; Zhang et al. 2019; Zhu, Cai, et al. 2020). This study adopted network pharmacological results to pre-clinical experiments, starting from the material basis of AOB, analyzing and exploring the mechanism of action of AOB in the treatment for AS, and at the same time providing a certain theoretical basis for clinical application and follow-up research.

Materials and methods

Collection of chemical components for AOB and screening of active compounds

We used Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, https://old.tcmsp-e.com/tcmsp.php) to screen the active ingredients of each herb (Ru et al. 2014). We then identified compounds with oral bioavailability (OB) ≥ 30% (Xu, Zhang, et al. 2012) and drug-likeness (DL) ≥ 0.18 (Jia et al. 2020) in AOB as compounds with pharmacological activity, based on absorption, distribution, metabolism and excretion (ADME) characteristics of the drugs in the body. After the preliminary screening of compounds, the PubChem database (https://pubchem.ncbi.nlm.nih.gov) was used to confirm their molecular structure and name, to improve the credibility of screening results. The identified molecules were entered into the SwissTargetPrediction website (swisstargetprediction.ch) to find the protein targets of the active compounds, and related targets were added based on published literature reports. Then, the screened protein targets were unified in the Uniprot protein database (https://www.uniprot.org) for specification and protein-gene docking for further prediction and analysis.

Prediction of potential targets of AOB for treatment

GeneCards is a searchable comprehensive database that automatically integrates gene-centric data from approximately 150 web sources, including genomics, transcriptomics, proteomics, genetics, clinical and functional information (Rebhan et al. 1997). With ‘Atherosclerosis’ as the keyword, relevant gene target information was searched in the GeneCards database (https://www.genecards.org) (Rebhan et al. 1997), and potential genes were supplemented using the TTD database (http://db.idrblab.net/ttd/) (Hamosh et al. 2005). When the number of targets is too large, the Score value in the Genecards database can be used for screening. The larger the score value, the closer the relationship between the target and the disease. The median of the Score value is used as the screening value. When there is too much data, multiple screening can be performed to obtain AS-related targets. The intersection of drug component-related targets and AS targets was operated by Venny2.1 (https://bioinfogp.cnb.csic.es/tools/venny/).

Construction of an active compound-disease-target network

Upload the intersection of targets to the STRING11.0 database (https://string-db.org) (Szklarczyk et al. 2017) to construct a protein-protein interaction (PPI) network model, set the biological species to ‘Homo sapiens’, and set ‘highest confidence’ > 0.9. The PPI network was obtained by screening, and the PPI network was further clustered by Cytoscape_v3.8.2 (Shannon et al. 2003) to obtain potential protein functional modules. The core targets are selected according to the comprehensive ranking of node connectivity (degree), node closeness (closeness) and node betweenness (betweenness).

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis

The core target genes obtained in the above steps were uploaded to the Metascape platform (http://metascape.org) (Zhou et al. 2019), and a threshold of p < 0.01 was set. The main biological processes and metabolic pathways were analyzed, including the KEGG pathway, GO biological process, Cell composition, and molecular function enrichment analysis. Then the data were saved and the results were visualized. Finally, a visualization network of ‘prescriptions – traditional Chinese medicine – chemical components – core targets – key pathways’ was built.

The preparation process of AOB

Alismatis rhizoma, Atractylodis macrocephalae rhizoma, and Pyrolae herba were purchased from Tong Ren Tang, Beijing. Alismatis rhizoma (100 ± 5 g), Atractylodis macrocephalae rhizoma (100 ± 5 g) and Pyrolae herba (50 ± 2 g) were placed into 5 L water to soak for 1 h. After soaking, we put the drug at 100 °C for condensation reflux extraction for 2 h and then recycled the medicine liquid and performed rotary evaporation and concentration at a constant temperature of 55 °C. After the concentrated medicinal liquid was recovered, it was freeze-dried at −60 °C and made into freeze-dried powder. The yield is 33.7%, which meets the requirements of drug preparation. The HPLC profile is shown in Figure 1. Both the peak area and concentration of Alisol A and Alisol B 23-monoacetate in AOB are presented in Table 1.

Figure 1.

Figure 1.

The HPLC fingerprint of AOB (A), benchmark sample (B).

Table 1.

Components of AOB observed by HPLC.

Both peak area and concentration of Alisol A and Alisol B 23-monoacetate in AOB by HPLC analysis
Sample Concentration (mg/mL) Peak area
Alisol A 0.353 8,586,146
AOB 0.139 3,383,560
Alisol B 23-monoacetate 0.773 8,459,846
AOB 0.396 4,334,478

Animals

Ten male C57BL/6J mice and 20 male APOE−/− mice were purchased from Changzhou Cavens Laboratory Animal Co., Ltd. (NO. SCXK (SU)-2016-0010). The weight of the mice was 22-24 g and the age of the mice was 4 weeks. All mice were acclimated to a standard rearing environment (Temperature: 18–22 °C, Humidity: 50–60%) for 1 week with 5 mice per cage before experiments carried out. All animal experiments were approved by the Animal Ethics Committee of Nanjing University of Chinese Medicine (NO. 202106A024). All animal experiments complied with animal ethics and all experiments were double-blind.

High-fat diet model (HFD), experimental design and AOB treatment

We designed three groups including control (CON), model (MOD), Alisma orientalis beverage (AOB). In the study, C57BL/6J mice were assigned to the control group, which were fed with a normal mouse diet; and APOE−/− were randomly assigned to the MOD group and AOB group. Mice in the MOD group were fed with a high-fat diet model for 8 weeks; AOB (3.8 g/kg, i.g.) was given daily while feeding with a high-fat diet for 8 weeks in the AOB group. All drugs followed the dosage of the Chinese Pharmacopoeia and did not impair liver and kidney function in mice after 8 weeks of AOB (Table 2).

Table 2.

Liver function and renal function after AOB treatment for 8 weeks.

  Reference range
Groups Liver function
Renal function
ALT
(10.06–96.47 U/L)
AST
(36.31–235.48 U/L)
CR
(10.91–85.09 Umol/L)
BUN
(10.81–34.74 mg/dL)
CON (Mean ± SD) 43.549 ± 1.655 209.314 ± 4.336 26.314 ± 4.824 22.624 ± 1.106
MOD (Mean ± SD) 74.953 ± 15.255 171.605 ± 8.584 56.165 ± 4.455 15.499 ± 1.232
AOB (Mean ± SD) 59.124 ± 9.392 192.546 ± 9.641 45.406 ± 9.737 13.734 ± 2.006
AOD (Mean ± SD) 68.734 ± 16.558 213.935 ± 18.442 51.105 ± 6.759 14.733 ± 2.396

Analysis of blood lipids

All mice were anesthetized by using pentobarbital sodium (45 mg/kg; i.p.). After anesthetization, blood was collected by enucleating the eyeball. The collected blood was centrifuged at low speed (3000 rpm) at 4 °C for 10 min, then collected supernatant and stored in −20 °C. High-density lipoprotein cholesterol (HDL-C) and triglycerides (TG), the serum levels of CHO and low-density lipoprotein cholesterol (LDL-C) were measured using a Chemray 240 automatic biochemical analyzer (Wuhan Servicebio Technology, Co., Ltd., China). All experiments were performed as described by the manufacturer.

Aortic plaque analysis

The heart and the aortic arch were taken out at a low temperature of 4 °C, and the residual blood was washed with 0.01 M PBS. Tissues were placed in a cryostat (Leica, Germany) and serial sections (10 μm) from the aortic sinus to the aortic arch were made from the aortic root according to anatomical markers for histological examination of atherosclerotic aortic sinus lesions. Oil red O staining (ORO) and HE staining were subsequently performed. The plaque area was analyzed using Image Pro Plus 6.0 (Image analysis software, Media Cybernetics, Rockville, MD, USA).

Western blot

The mouse liver tissue (100 mg/kg) was taken out at a low temperature and placed in a lysate for sufficient grinding to a particle-free state, followed by the addition of protease inhibitors. We then centrifuged for 10 min and took the supernatant for a protein concentration test (protein concentration by BCA method). Moreover, the samples were equalized according to the protein concentration and cooked at 100 °C for 5 min with Loading buffer added until the protein was stable. After the target protein was separated by gel electrophoresis (80 v, 90 min), which was transferred to the PVDF membrane under constant flow (300 mA, 60 min, 4 °C). After 18 h of primary antibody including pPI3K (1:1000), PI3K (1:1000), pAKT (1:1000), AKT (1:1000), SREBP-1 (1:1000), GAPDH (1:5000) incubation at 4 °C, we made 2 h of secondary antibody (IgG-Rabbit, 1:4000) incubation at room temperature (20–26 °C), ECL imaging was performed. Visualization of the blot was performed with the chemiluminescent substrate SuperSignal West Pico (Thermo Fisher Science Inc.) and displayed as density relative to GAPDH. Experiments were performed at least 3 times.

Statistical analysis

All data were shown in the form of mean ± SEM. One-way ANOVA was used with the honestly important difference from Tukey or the post-hoc test from Dunnett. For all statistical tests, GraphPad Prism 8.0 was used, and one-way ANOVA was used in three groups. p < 0.05 was considered statistically significant.

Results

Identification of potential action targets of AOB

We collected 137 compounds in AOB from the TCMSP database, 46 of which belonged to Alismatis rhizoma, 55 belonged to Atractylodis macrocephalae rhizoma and 36 belonged to Pyrolae herba. After screening by ADME, a total of 7 active ingredients of Alismatis rhizoma, 4 from Atractylodis macrocephalae rhizoma, 5 from Pyrolae herba, and 1 common active ingredient from Alismatis rhizoma and Pyrolae herba were obtained, including Alisma alcohol, kaempferol, quercetin, gallic acid, atractylodes lactone, etc. (Table 3). Furthermore, we collected the targets of 17 active compounds in AOB from the TCMSP. After the integration of UniProt database entries and the deletion of duplicates, 601 targets were obtained (Table 4).

Table 3.

The active compounds of AOB.

Alisma orientalis beverage
Mol ID Herb name Molecule name OB (%) DL
MOL000359 Alisma orientale (Sam.) Juz.
Pyrola calliantha H. Andres
β-Sitosterol 36.91 0.75
MOL000831 Alisma orientale (Sam.) Juz. Alisol B monoacetate 35.58 0.81
MOL000832 Alisma orientale (Sam.) Juz. Alisol B 23-acetate 32.52 0.82
MOL000849 Alisma orientale (Sam.) Juz. 16β-Methoxyalisol B monoacetate 32.43 0.77
MOL000853 Alisma orientale (Sam.) Juz. Alisol B 36.76 0.82
MOL000854 Alisma orientale (Sam.) Juz. Alisol C 32.7 0.82
MOL000856 Alisma orientale (Sam.) Juz. Alisol C monoacetate 33.06 0.83
MOL002464 Alisma orientale (Sam.) Juz. 1-Monolinolein 37.18 0.3
MOL000033 Atractylodes macrocephala Koidz. (24S)-24-Propylcholesta-5-ene-3beta-ol 36.23 0.78
MOL000028 Atractylodes macrocephala Koidz. α-Amyrin 39.51 0.76
MOL000049 Atractylodes macrocephala Koidz. 3β-Acetoxyatractylone 54.07 0.22
MOL000072 Atractylodes macrocephala Koidz. 8β-Ethoxy atractylenolide III 35.95 0.21
MOL000422 Pyrola calliantha H. Andres Kaempferol 41.88 0.24
MOL000552 Pyrola calliantha H. Andres 5,2′-Dihydroxy-6,7,8-trimethoxyflavone 31.71 0.35
MOL000553 Pyrola calliantha H. Andres (−)-Chimonanthine 38 0.66
MOL000569 Pyrola calliantha H. Andres Digallate 61.85 0.26
MOL000098 Pyrola calliantha H. Andres Quercetin 46.43 0.28

Table 4.

The targets of active compounds in AOB.

Targets of AOB
NPC1L1 HTR2A PLA2G2A IL2 IDH1 ABCC1 CCR1 HSD17B1
NR1H3 HTR2C CSNK2A1 UCK2 PDE3A AHR CNR1 TYMS
RORC MMP1 GSK3B NCS1 FNTA ABCB1 C5AR1 AKR1C1
HMGCR MAPK14 TTPA DPEP1 NAMPT CYP1B1 DUT CHIT1
SHBG PGA5 FABP6 HCK GPR88 ABCG2 JAK3 CTSB
CYP51A1 FNTB PDK2 IMPDH2 HSD17B2 ADORA1 KDR SRC
CYP17A1 PGGT1B PPARA YARS1 FKBP1A CA4 DRD3 GSR
CYP19A1 OXTR ADK CTSF PDE2A ALOX15 MDM2 AURKA
SREBF2 IKBKB DHFR CSK PYGM ALOX12 AVPR1A CSNK1G2
AR TTR SYK RARB MAST3 PTPRS PDE10A NOS3
RORA BMP2 FGFR1 AMY2A CETP ADORA2A EDNRB SORD
ESR1 CFB EPHX2 RNASE3 CCNB3 CCNB3 MC4R LGALS7
ESR2 MAPK10 CTNNA1 AMY1A CASR GPR35 MC1R LGALS7B
PTPN1 THRB TRAPPC3 AMY1B GABRB3 DAPK1 HSD17B3 DPP4
CYP2C19 ALB ELANE AMY1C PSENEN MPG TK2 ADAM17
SLC6A2 PGR CTSK BHMT NCSTN SLC22A12 MC5R APCS
ACHE CA2 ITK ABL1 APH1A TNKS2 MC3R ADH1B
SERPINA6 MAPKAPK2 PCK1 B3GAT1 PSEN1 TNKS AVPR2 HSP90AA1
G6PD EPHB4 MET MMP9 APH1B MPO CNR2 F10
BCHE ITGAL GSTA1 RXRB GABRA3 PTK2 MAP3K14 PNPO
SLC6A4 PIM1 FABP7 ACE GABRG2 CA3 ADA CFD
CHRM2 PPIA PAH RAB11A CDK1 CA6 DRD4 ANG
NR1I3 CASP7 PDE3B C1S CCNB1 PKN1 NR3C2 SEC14L2
NR1H2 ANXA5 PRKACA DTYMK CCNB2 CA14 NR3C1 HSPA8
DHCR7 MAOB AMD1 CD1A PDGFRB NEK2 CCKBR FABP4
PTGER1 AKR1C2 MMP12 JAK2 ROCK2 CXCR1 CHUK CTSV
PTGER2 MAPK8 SOD2 GPI LIMK2 CAMK2B KCNA5 LCK
VDR TREM1 FGFR2 ACADM CDK9 AKT1 SLC6A3 C1R
TBXAS1 GSTP1 MMP8 GCK IRAK4 NEK6 SMO MTHFD1
PTGES MMP3 PPP5C EPHA2 LRRK2 CA5A FAAH IGLV2-8
PPARD AKR1B1 CYP2C9 AKT2 AURKAIP1 AXL MAPK1 NQO1
CES2 CES1 ADH5 GRB2 NPY5R NUAK1 F2RL1 PARP1
HSD11B1 GC CTSG TEK TK1 AKR1C4 REN ESRRG
SQLE HSD17B11 ALDH2 SETD7 AMPD2 CA13 JAK1 HDAC8
PTPN6 KIF11 CDA PAPSS1 FASN AKR1A1 MCHR1 PLA2G10
PTPN2 CHEK1 AZGP1 ALAD TRPV1 APP F2 ADH1C
GLRA1 PDE4B PLAU GSTT2B S1PR3 CD38 STS RXRA
NOS2 GBA CBR1 TGFB2 S1PR1 TOP1 ALK PLK1
PPARG PIK3CG ATOX1 LGALS3 ALOX5 CFTR CSF1R BCAT2
UGT2B7 PNP CA1 NMNAT1 LPAR6 GRK6 DRD1 PDPK1
POLB MMP13 FHIT RAC2 LPAR5 CCNB1 SCD HTR3A
PRKCA FKBP1A RBP4 LTA4H ENPP2 ODC1 ACP1 NOS1
PRKCD APOA2 FECH AGXT SLC8A1 ADORA3 AKR1B10 ADRA2A
PRKCQ CMA1 SHMT1 PAK6 CTSL MCL1 CD81 ADRA2C
PTGS2 CCNA2 ISG20 PIK3R1 PREP PLG ARG1 HRH2
FNTA CASP3 BLVRB CLK1 S1PR5 PLA2G7 FOLH1 ADRA2B
VAV1 PDE5A AHCY RHOA S1PR4 MAPT GSTM1 CXCR4
PGGT1B DHODH FABP3 MAN1B1 ACACB TOP2A NT5M SCARB1
F2R CDK5R1 SULT1E1 PPP1CC PIK3CD APEX1 TPSB2 SCN4A
TACR1 CDK2 SULT2B1 ARSA PIM3 GUSB FKBP3 CXCR3
PRKCB MIF DCK MME PFKFB3 HSP90AB1 INSR SIGMAR1
PDE4D TNNC1 FABP5 PLEKHA4 AURKB RET WARS1 PRCP
METAP2 TGFBR1 ABO F11 TYK2 NAE1 RFK ADRA1D
P2RX3 SULT2A1 REG1A IMPDH1 F9 PCSK7 PROCR CTSC
TRPV4 EGFR NR1I2 HRAS KCNH2 DRD5 FGG KHK
PRKCG PTPN11 ERBB4 PCTP ERBB2 HRH4 MAOA ARHGDIA
PRKCE NR1H4 ME2 TPH1 PTK6 HTR1E PLA2G1B SSTR4
PRKCH WAS XIAP HAGH FDFT1 HTR5A RPS6KA5 SSTR1
BACE2 BACE1 F7 BCL2L1 TERT OPRL1 GLRA2 CACNA1G
CTSD AKR1C3 KYAT1 CASP1 FABP1 OPRK1 ATP12A PIM2
PER2 GLO1 HNF4G THRA ZAP70 MMP2 PYGL HNMT
MTAP IGF1R RARG ESRRA CDK6 SDS IGF1 NQO2
MAP2K1 PSAP GM2A MMP7 S100A9 CYP2C8 CTSS BST1
HK1 RARA LCN2 PNMT PGF SERPINA1 ACP3 ALDOA
HPN CRABP2 TGM3 CCNT1 CCL5 C8G BIRC7 PADI4
TPI1 TTL CDK5R1 CACNA1B PSEN2 CA12 CA9 CDC25A
CDC25B RIPK3 RIPK2 CDK5 PSENEN NCSTN APH1A PSEN1
NOX4 XDH TYR FLT3 CA7 HEXB FDPS GNPDA1
STAT1 LGALS2 CDK7 KIT GSTO1 PITPNA EIF4E MTNR1A
MTNR1B ACKR3 HSD11B2 SCN9A PSEN2 OPRM1 OPRD1 CCNC
CDK8 IL6ST MTOR PIK3CA PDGFRB NTRK1 PAK1 PIK3CB
HCRTR2 HCRTR1 KCNK3 GYS1 POLA1 FYN PDGFRA EPHB3
GSK3A VHL EZH2 FAP P2RX7 CHRNB3 HASPIN KCNN4
SSTR3 MAP3K8 NOD1 NOD2 TRPM8 PHLPP2 SLC6A1 DRD2
CCNA1 CHRNA6 CHRNB2 CHRNA3 SERPINE1 TUBB1 FUT7 KDM4E
MYLK              

We collected 4481 AS targets from the Genecards database. The median of the Score value was used as the screening value, so the target with a Score > 2.77 was set as the potential target of AS. We combined with OMIM, TTD, and DRUGBANK databases to supplement relevant targets, and deleted duplicate values after merging, and finally obtaines 1128 dyslipidemia-related targets. We took the intersection of the screened drug active ingredient targets and AS targets, and drew Venn diagrams through Venny2.1 to obtain 171 common targets of AOB and AS (Figure 2 and Table 5).

Figure 2.

Figure 2.

Targets screening involved in AOB for the treatment of AS. Venn diagram of disease targets.

Table 5.

The 171 common targets of AOB and AS.

Common targets of AOB and AS
NPC1L1 PTGS2 JAK1 GC NOS3 SOD2 CSK BCL2L1
NR1H3 PGGT1B F2 GBA DPP4 MMP8 BHMT CASP1
HMGCR F2R CSF1R PIK3CG ADAM17 CYP2C9 MMP9 STAT1
SHBG PDE4D HTR2A MMP13 HSP90AA1 ALDH2 ACE LGALS2
CYP19A1 PRKCE MMP1 APOA2 F10 AHCY C1S GSTO1
SREBF2 CTSD MAPK14 CMA1 FABP4 FABP3 JAK2 PITPNA
AR CCR1 IKBKB CASP3 NQO1 REG1A AKT2 PSEN2
ESR1 CNR1 TTR PDE5A PARP1 NR1I2 TEK IL6ST
ESR2 C5AR1 MAPK10 MIF PLA2G2A F7 TGFB2 MTOR
CYP2C19 KDR ALB TGFBR1 GSK3B MMP2 LGALS3 PIK3CA
ACHE EDNRB ITGAL EGFR TTPA IGF1 LTA4H PIK3CB
BCHE NR3C2 ANXA5 PTPN11 PPARA MMP7 AGXT NAMPT
SLC6A4 NR3C1 MAOB NR1H4 SYK S100A9 PIK3R1 CETP
NR1H2 CHUK MAPK8 BACE1 EPHX2 LCN2 RHOA CASR
PPARD SLC6A3 GSTP1 CHIT1 CTNNA1 PGF ARSA PSEN1
NOS2 MAPK1 MMP3 SRC ELANE CCL5 MME LRRK2
PPARG REN CES1 GSR MMP12 IL2 F11 ALOX5
POLB AKT1 APP TOP1 CFTR PLG PLA2G7 MAPT
HTR3A NOS1 CXCR4 SCARB1 CXCR3 KHK DRD2 SERPINE1
CTSL F9 KCNH2 FDFT1 ARG1 GSTM1 PROCR FGG
PLA2G1B NOX4 XDH ABCC1 ABCB1 ALOX15 ADORA2A MPO
PTK2 APEX1 MYLK          

The potential targets of AOB for the treatment of AS

To comprehensively elucidate the possible mechanism of AOB in the treatment of AS, 171 AOB anti-AS target gene names were imported into the STRING database to construct a PPI network. The required interaction score was 0.9 and the disconnected node network was hidden to draw a PPI network graph (Figure 3(A)). To achieve better visualization and identify core targets, we build a network using Cytoscape based on target degrees. With this network, core targets were obtained: PIK3R1, AKT1, PIK3CA, MAPK1, PTPN11, EGFR and MAPK4 (Figure 3(B) and Table 6). These targets may be considered as primary targets of action for AOB for AS treatment, and their identification suggests that AOB treats AS through multiple potential targets.

Figure 3.

Figure 3.

The potential targets of AOB for the treatment of AS. (A) The PPI network was constructed by the STRING database. (B) Drawing the PPI core network with Cytoscape 3.8.2 for visual display.

Table 6.

The top 10 targets of the PPI Network.

Target Betweenness centrality Closeness centrality Degree
HSP90AA1 0.166047284 0.432343234 60
PIK3R1 0.081179043 0.411949686 58
SRC 0.133317904 0.433774834 56
AKT1 0.074995392 0.401840491 50
PIK3CA 0.029786286 0.384164223 50
MAPK1 0.109227209 0.413249211 48
PTPN11 0.023755181 0.388724036 46
EGFR 0.032745575 0.388724036 42
RHOA 0.058774436 0.389880952 40
MAPK14 0.039497188 0.374285714 32

GO and KEGG enrichment analysis for identification of the pathway mechanisms of AOB

The Metascape data platform was used to analyze the signal pathway of the related targets in the regulation of AS by AOB. AOB was mainly involved in the biological processes including regulation of cell adhesion, wound healing, positive regulation of protein phosphorylation, positive regulation of cell migration, etc. The main cellular components involved include membrane raft, the extrinsic component of the membrane, phosphatidylinositol 3-kinase complex, focal adhesion, etc. GO molecular functions of AOB involved include phosphotransferase activity, alcohol group as acceptor, protein kinase activity, kinase activity, kinase binding, protein kinase binding, phosphatase binding, transmembrane receptor protein tyrosine kinase activity, protein tyrosine kinase activity, transmembrane receptor protein kinase activity, protein phosphatase binding. The pathways involved mainly include cancer pathways, PI3K-AKT signaling pathway, EGFR tyrosine kinase inhibitor resistance, fluid shear stress and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, hepatitis B, etc. (Table 7). The top 10 significantly enriched (p < 0.01) terms in BP, CC and MF of GO analysis were selected (Figure 4(A)). The top 20 pathways with significant enrichment (p < 0.01) were selected (Figure 4(B)).

Table 7.

KEGG enrichment analysis results.

Category GO Description LogP Count
KEGG pathway hsa05200 Pathways in cancer −39.55364346 31
KEGG pathway hsa04151 PI3K-Akt signaling pathway −27.80028128 22
KEGG pathway hsa01522 Endocrine resistance −27.56991838 16
KEGG pathway hsa05205 Proteoglycans in cancer −27.25628276 19
KEGG pathway hsa01521 EGFR tyrosine kinase inhibitor resistance −26.73995834 15
KEGG pathway hsa05215 Prostate cancer −24.94056688 15
KEGG pathway hsa05418 Fluid shear stress and atherosclerosis −24.3778864 16
KEGG pathway hsa04933 AGE-RAGE signaling pathway in diabetic complications −22.50156695 14
KEGG pathway hsa04917 Prolactin signaling pathway −22.49866763 13
KEGG pathway hsa05212 Pancreatic cancer −22.03626506 13
KEGG pathway hsa05160 Hepatitis C −21.49278225 15
KEGG pathway hsa05161 Hepatitis B −21.18256658 15
KEGG pathway hsa04062 Chemokine signaling pathway −20.30080047 15
KEGG pathway hsa05162 Measles −20.02945253 14
KEGG pathway hsa04630 Jak-STAT signaling pathway −19.53523346 14
KEGG pathway hsa04611 Platelet activation −19.29312092 13
KEGG pathway hsa04012 ErbB signaling pathway −19.13592737 12
KEGG pathway hsa05210 Colorectal cancer −19.13592737 12
KEGG pathway hsa04370 VEGF signaling pathway −18.98104023 11
KEGG pathway hsa04014 Ras signaling pathway −18.65825684 15
KEGG pathway hsa04915 Estrogen signaling pathway −18.65571774 13
KEGG pathway hsa04931 insulin resistance −18.13117073 12
KEGG pathway hsa04510 Focal adhesion −18.11591861 14
KEGG pathway hsa04668 TNF signaling pathway −18.0360695 12
KEGG pathway hsa04072 Phospholipase D signaling pathway −17.9771753 13
KEGG pathway hsa04066 HIF-1 signaling pathway −17.85103726 12
KEGG pathway hsa05164 Influenza A −17.34180853 13
KEGG pathway hsa04380 Osteoclast differentiation −17.0538665 12
KEGG pathway hsa04550 Signaling pathways regulating pluripotency of stem cells −16.50259738 12
KEGG pathway hsa05142 Chagas disease (American trypanosomiasis) −16.46447588 11
KEGG pathway hsa04660 T cell receptor signaling pathway −16.19441652 11
KEGG pathway hsa04015 Rap1 signaling pathway −16.05447112 13
KEGG pathway hsa04664 Fc epsilon RI signaling pathway −15.92581654 10
KEGG pathway hsa04670 Leukocyte transendothelial migration −15.8570988 11
KEGG pathway hsa04919 thyroid hormone signaling pathway −15.77645797 11
KEGG pathway hsa04722 Neurotrophin signaling pathway −15.73665629 11
KEGG pathway hsa04071 Sphingolipid signaling pathway −15.61925748 11
KEGG pathway hsa04068 foxo signaling pathway −15.24808488 11
KEGG pathway hsa05222 Small cell lung cancer −14.90283077 10
KEGG pathway hsa04210 Apoptosis −14.77385153 11
KEGG pathway hsa04914 Progesterone-mediated oocyte maturation −14.72482596 10
KEGG pathway hsa05213 Endometrial cancer −14.6007413 9
KEGG pathway hsa04150 mTOR signaling pathway −14.55382048 11
KEGG pathway hsa05169 Epstein-Barr virus infection −14.52462989 13
KEGG pathway hsa05224 Breast cancer −14.43264667 11
KEGG pathway hsa0460 Toll-like receptor signaling pathway −14.38982393 10
KEGG pathway hsa05214 Glioma −13.97694069 9
KEGG pathway hsa04662 B cell receptor signaling pathway −13.8230983 9
KEGG pathway hsa05220 Chronic myeloid leukemia −13.77316427 9
KEGG pathway hsa05231 Choline metabolism in cancer −12.69949295 9
KEGG pathway hsa05221 Acute myeloid leukemia −12.42231383 8
KEGG pathway hsa01524 Platinum drug resistance −12.27052195 8
KEGG pathway hsa05166 HTLV-I infection −12.24154346 11
KEGG pathway hsa05230 Central carbon metabolism in cancer −12.03171822 8
KEGG pathway hsa04360 Axon guidance −11.88732319 10
KEGG pathway hsa04140 Regulation of autophagy −11.8636379 9
KEGG pathway hsa05218 Melanoma −11.80864445 8
KEGG pathway hsa05203 Viral carcinogenesis −11.56800581 10
KEGG pathway hsa04910 Insulin signaling pathway −11.2303346 9
KEGG pathway hsa04930 Type II diabetes mellitus −11.08590908 7
KEGG pathway hsa04810 Regulation of actin cytoskeleton −11.08538891 10
KEGG pathway hsa04750 inflammatory mediator regulation of trp channels −11.00553138 8
KEGG pathway hsa04666 Fc gamma R-mediated phagocytosis −10.93815983 8
KEGG pathway hsa04932 Non-alcoholic fatty liver disease (NAFLD) −10.89148672 9
KEGG pathway hsa04213 Longevity regulating pathway – multiple species −10.87448477 7
KEGG pathway hsa04725 Cholinergic synapse −10.65068367 8
KEGG pathway hsa05223 Non-small cell lung cancer −10.49206217 7
KEGG pathway hsa05211 Renal cell carcinoma −9.997610762 7
KEGG pathway hsa05100 Bacterial invasion of epithelial cells −9.88593232 7
KEGG pathway hsa04024 cAMP signaling pathway −9.763259543 9
KEGG pathway hsa04211 Longevity regulating pathway −9.118717848 7
KEGG pathway hsa04152 AMPK signaling pathway −8.60443639 7
KEGG pathway hsa04650 Natural killer cell mediated cytotoxicity −8.18812509 7
KEGG pathway hsa04960 Aldosterone-regulated sodium reabsorption −8.16782521 5
KEGG pathway hsa05146 Amoebiasis −7.517592689 6
KEGG pathway hsa04973 Carbohydrate digestion and absorption −7.478108164 5
KEGG pathway hsa04923 Regulation of lipolysis in adipocytes −7.277689618 5
KEGG pathway hsa00562 Inositol phosphate metabolism −3.494032868 3
KEGG pathway hsa04070 Phosphatidylinositol signaling system −3.124304392 3
KEGG pathway hsa05145 Toxoplasmosis −21.98392135 14
KEGG pathway hsa05152 Tuberculosis −17.03489964 13
KEGG pathway hsa04659 Th17 cell differentiation −16.23835335 11
KEGG pathway hsa04658 Th1 and Th2 cell differentiation −11.21614661 8
KEGG pathway hsa04621 NOD-like receptor signaling pathway −10.51759803 9
KEGG pathway hsa05140 Leishmania infection −8.5243026 6
KEGG pathway hsa05168 Herpes simplex infection −7.518063175 7
KEGG pathway hsa04657 IL-17 signaling pathway −15.0886455 10
KEGG pathway hsa04010 MAPK signaling pathway −12.6446435 12
KEGG pathway hsa05120 Epithelial cell signaling in Helicobacter pylori infection −12.37095867 8
KEGG pathway hsa05133 Pertussis −8.248605395 6
KEGG pathway hsa05131 Shigellosis −6.926342175 5
KEGG pathway hsa04728 Dopaminergic synapse −5.489969573 5
KEGG pathway hsa05132 Salmonella infection −4.656866744 4
KEGG pathway hsa04622 RIG-I-like receptor signaling pathway −3.580079392 3
KEGG pathway hsa04723 Retrograde endocannabinoid signaling −2.523302304 3
KEGG pathway hsa04920 Adipocytokine signaling pathway −12.3203688 8
KEGG pathway hsa04922 Glucagon signaling pathway −3.076284659 3
KEGG pathway hsa05219 Bladder cancer −9.890157853 6
KEGG pathway hsa04912 GnRH signaling pathway −7.720652493 6
KEGG pathway hsa04520 Adherens junction −6.543089003 5
KEGG pathway hsa04921 Oxytocin signaling pathway −6.462524366 6
KEGG pathway hsa04540 Gap junction −3.295118179 3
KEGG pathway hsa04371 Apelin signaling pathway −9.815534753 8
KEGG pathway hsa04022 cGMP-PKG signaling pathway −7.718646373 7
KEGG pathway hsa04261 Adrenergic signaling in cardiomyocytes −5.261253948 5
KEGG pathway hsa05206 MicroRNAs in cancer −7.279201986 8
KEGG pathway hsa04270 Vascular smooth muscle contraction −2.767307962 3
KEGG pathway hsa04726 Serotonergic synapse −7.241068175 6
KEGG pathway hsa00590 Arachidonic acid metabolism −3.751970004 3
KEGG pathway hsa05010 Alzheimer’s disease −4.942122898 5
KEGG pathway hsa05202 Transcriptional misregulation in cancer −4.573319263 5
KEGG pathway hsa04064 NF-kappa B signaling pathway −4.45368227 4
KEGG pathway hsa04215 Apoptosis – multiple species −4.598201729 3
KEGG pathway hsa05014 Amyotrophic lateral sclerosis (ALS) −3.881447695 3
KEGG pathway hsa04137 Mitophagy – animal −3.67260526 3
KEGG pathway hsa04115 p53 signaling pathway −3.461237887 3
KEGG pathway hsa04144 Endocytosis −3.988916403 5
KEGG pathway hsa04060 Cytokine-cytokine receptor interaction −3.603564725 5
KEGG pathway hsa00330 Arginine and proline metabolism −3.731646229 3
KEGG pathway hsa04020 Calcium signaling pathway −3.366224834 4
KEGG pathway hsa04530 Tight junction −3.589422319 4
KEGG pathway hsa04310 Wnt signaling pathway −2.50874465 3
KEGG pathway hsa05323 Rheumatoid arthritis −3.17432923 3
KEGG pathway hsa04114 Oocyte meiosis −2.832488553 3

Figure 4.

Figure 4.

GO and KEGG enrichment analysis for identification of the pathway mechanisms of AOB. (A) The top 10 significantly enriched (p < 0.01) terms in BP, CC and MF of GO analysis were selected. (B) The top 20 pathways with significant enrichment (p < 0.01) were selected.

Network-based revelation of Compound-Disease-Pathway-Target network correlations

Combined with the above analysis results, the connection between traditional Chinese medicine, disease, pathways and targets was established. CytoScape3.8.2 was used to construct a Compound-Disease-Pathway-Target network (Figure 5). With the use of the built-in NetworkAnalyzer of CytoScape3.8.2, the network topology parameters of AOB treatment AS were analyzed, and the core components and core role targets were obtained. According to network analysis, 3 main components in the AOB treatment of AS: 16β-methoxyalisol B monoacetate (degree = 36), 3β-acetoxyatractylone (degree = 23), and 5, 2′-dihydroxy-6,7,8-trimethoxyflavone (degree = 19) (Table 8). Therefore, these compounds were regarded as the potential bioactive compounds of AOB against AS. Then, the top 10 core targets were selected according to the comprehensive ranking of degree, closeness and betweenness (Table 9). Interestingly, according to the network analysis results, the PIK3 family and AKT are the core targets of AOB in the treatment of AS, which is consistent with the previous PPI analysis results.

Figure 5.

Figure 5.

Compound-Disease-Pathway-Target Network. The orange hexagons represent AOB, the yellow hexagons represent AS, the green circles represent three traditional Chinese medicines, the blue diamonds around the green circles are the main components of the medicine, the red arrows represent the pathways, and the outermost blue circles are the targets. The darker the color, the more important the node is.

Table 8.

Main components in the AOB treatment of AS.

Herb name Mol ID Betweenness centrality Closeness centrality Degree
Alisma orientale (Sam.) Juz. MOL000849 0.120442743 0.539393939 36
Atractylodes macrocephala Koidz. MOL000049 0.056925762 0.465968586 23
Pyrola calliantha H. Andres MOL000552 0.039327035 0.447236181 19

Table 9.

The top 10 targets of the Compound-Disease-Pathway-Target Network.

Target Betweenness centrality Closeness centrality Degree
PIK3R1 0.025841461 0.497206704 18
MAPK1 0.031190839 0.486338798 18
PIK3CA 0.018435115 0.475935829 17
PIK3CB 0.018435115 0.475935829 17
AKT1 0.020792845 0.486338798 17
AKT2 0.015704295 0.470899471 16
MAPK14 0.035347078 0.470899471 15
MAPK8 0.016521855 0.470899471 15
SRC 0.023938519 0.486338798 15
GSK3B 0.022214607 0.481081081 14

AOB reversed the aortic plaque area of as in APOE−/− mice stimulated with a high-fat diet

We performed an AS model with a high-fat diet (HFD). We found that C57BL/6J mice fed with a normal diet for 8 weeks showed no change in arterial plaque area and APOE−/− mice fed with HFD for 8 weeks showed a significant increase in arterial plaque area, which was compared with C57BL/6J mice fed with normal diet (Figure 5). However, after AOB treatment for 8 weeks, the arterial plaque area was significantly reversed both in ORO and HE staining (Figure 6; One-way ANOVA, ORO, F (2, 26) = 62.35, p < 0.001; HE, F (2, 26) = 86.91, p < 0.001). These data suggested that AOB had a potential effect to treat AS.

Figure 6.

Figure 6.

AOB reversed aortic plaque area of AS in APOE−/− mice. HE and ORO stained sections of aortic valve area in the control group, the model group and the AOB group. The atherosclerotic lesion area was quantitatively analyzed by Image J. Data show mean ± SEM values of 6 or more independent samples. # Represents comparison with the control group, ###represents p < 0.001; * represents comparison with the model group, *** represents p < 0.001.

AOB improved four indicators of blood lipids of as in APOE−/− mice stimulated with a high-fat diet

We then detected four indicators of blood lipids related to AS including TG, CHO, HDL and LDL. We found that TG, CHO, LDL were all increased after HFD feeding in APOE−/− mice for 8 weeks, which were all reversed obviously by AOB for 8 weeks [Figure 7, One-way ANOVA, TG, F (2, 17) = 85.17, p < 0.001; CHO, F (2, 17) = 59.30, p < 0.001; LDL, F (2, 17) = 19.20, p < 0.001]. Moreover, HDL in blood serum was decreased after HFD feeding in APOE−/− mice for 8 weeks, which was also reversed obviously by AOB for 8 weeks [Figure 7, One-way ANOVA, HDL, F (2, 17) = 76.44, p < 0.001].

Figure 7.

Figure 7.

AOB improved four indicators of blood lipid of AS in APOE−/− mice stimulated with high-fat diet. Data show mean ± SEM values of 6 independent samples. # Represents comparison with the control group, ### represents p < 0.001; * represents comparison with the model group, * represents p < 0.05, *** represents p < 0.001.

AOB alleviated as by regulating PI3K pathway

Based on our network pharmacological analysis, we chose the PI3K pathway to reveal the underlying mechanism of AOB treatment for AS. Then we measured the levels of PI3K/AKT/SREBP-1 in the liver. The results showed that HFD increased phosphorylated expressions of PI3K/AKT and expression of SREBP-1 in APOE−/− mice compared with C57BL/6J mice fed with a normal diet. Interestingly, AOB treatment for 8 weeks reversed all of them in the liver [Figure 8, One-way ANOVA, pPI3K/PI3K, F (2, 17) = 6.997, p = 0.0147; pAKT/AKT, F (2, 17) = 7.925, p = 0.0087; SREBP-1, F (2, 17) = 18.14, p < 0.001]. Although the expression of SREBP-2 in the liver was significantly increased by HFD, which was not altered after AOB treatment for 8 weeks [Figure 9, One-way ANOVA, F (2, 8) = 9.281 p = 0.0082].

Figure 8.

Figure 8.

AOB alleviated AS by regulating the PI3K pathway. The expression levels of the PI3K/AKT/SERBP-1 pathway proteins in each group were detected by western blots. The densitometric values of bands were quantitatively analyzed by Image J Densitometric values normalized to those in the model group and are presented as relative intensity. Data show mean ± SEM values of 6 independent samples. # Represents comparison with the control group, # represents p < 0.05, ## represents p < 0.01; * represents comparison with the model group, * represents p < 0.05, ** represents p < 0.01 *** represents p < 0.001.

Figure 9.

Figure 9.

The expression of SREBP-2 in liver after AOB treatment for 8 weeks. The densitometric values of bands were quantitatively analyzed by Image J Densitometric values normalized to those in the model group and are presented as relative intensity. Data show mean ± SEM values of 6 independent samples. # Represents comparison with the control group, ## represents p < 0.01.

Discussion

The relationship between the lipid metabolism pathway of AOB and AS was still unclear. In this study, we used network pharmacology combining with experiments to reveal the role of AOB in lipid metabolism, which played a major role in the treatment for AS. We found 3 main components in the AOB treatment of AS: 16β-methoxyalisol B monoacetate (degree = 36), 3β-acetoxyatractylone (degree = 23), and 5, 2′-dihydroxy-6,7,8-trimethoxyflavone (degree = 19) according to network analysis. Moreover, core targets were obtained: PIK3R1, AKT1, PIK3CA, MAPK1, PTPN11, EGFR and MAPK4, which might participate in the treatment of AS. In addition, the top 10 significantly enriched (p < 0.01) terms in BP, CC and MF of GO analysis were selected and the top 20 pathways with significantly enriched were selected, including the PI3K/AKT/SREBP-1 pathway. Based on the results, we used experiments to identified the therapeutic actions of AOB in AS via the PI3K/AKT/SREBP-1 pathway. The results showed that AOB reversed the aortic plaque area of AS, and improved main indicators of blood lipids related to AS and alleviated AS by regulating PI3K/AKT/SREBP-1 pathway in APOE−/− mice stimulated with HFD. Taken together, we firstly demonstrated that AOB was capable of ameliorating AS by regulating the PI3K/AKT/SREBP-1 pathway.

Although a previous study has identified the effects of AOB on the treatment for AS (Zhu, Zhai, et al. 2020), the underlying molecular mechanism was still unclear. The aortic plaque was significantly increased in AS-related diseases and had been shown to be closely related to high-fat diets (HFD) (Pan et al. 2022). In our study, we found a significant increase in the aortic plaque area in APOE−/− mice fed with HFD for 8 weeks, which was reversed by AOB after 8 weeks of continuous treatment. In addition, abnormal blood lipid indicators including TG, CHO, HDL and LDL, which contributed to AS (Jaquish et al. 1996; Barboza et al. 2016), were all relieved by AOB. To sum up, AOB has the effect of alleviating AS in APOE−/− mice stimulated with HFD.

Network pharmacological analysis showed that 3 main components, 16β-methoxyalisol B monoacetate, 3β-acetoxyatractylone, and 5,2′-dihydroxy-6,7,8-trimethoxyflavone, are associated with AOB treatment of AS. 16β-Methoxyalisol B monoacetate from Alismatis rhizoma has been identified to have an antibacterial effect (Jin et al. 2012) and the pathophysiology of bacterial is associated with the development of inflammation (Ge et al. 2022; Keir and Chalmers 2022), risk factors for atherosclerosis. Meanwhile, the component was proved to have an inhibitory effect on phosphorylation of the PI3K/Akt pathway (Xu, Zhao, et al. 2009). Moreover, 3β-acetoxyatractylone from Atractylodis macrocephalae rhizoma has been indicated to have treatment-related effects of AS (Chen et al. 2017; Li et al. 2018). In addition, 5,2′-dihydroxy-6,7,8-trimethoxyflavone from Pyrolae herba, a natural flavonoid, plays a role in lipid decreasing (Lin et al. 2022) and the progression of treatment of AS (Kimura et al. 2022; Liu et al. 2022), which also participate in anti-inflammation (Huang et al. 2022; Liu et al. 2022). Network pharmacological results were further identified in our experimental studies, which showed that AOB was capable of releiving AS by regulating the PI3K/AKT/SREBP-1 pathway.

Hypercholesterolemia is recognized as a major contributor to AS, and lowering blood cholesterol levels is an important means in the treatment of AS. Sterol-regulating element-binding proteins (SREBPs) are a family of transcription factors involved in the biosynthesis of cholesterol, fatty acids, and triglycerides (Moslehi and Hamidi-Zad 2018), consisting of SREBP-1 and SREBP-2. SREBP-1 is responsible for the synthesis of fatty acids and cholesterol, while SREBP-2 only regulates the synthesis of cholesterol (Jeon and Osborne 2012). Studies showed that SREBPs in the liver plays a catalytic role in AS by increasing lipid synthesis (Karasawa et al. 2011; Pérez-Belmonte et al. 2017), and inhibiting SREBP-1 led to lower serum cholesterol levels, further alleviating AS (Karasawa et al. 2011). PI3K/AKT is the upstream signaling pathway of SREBP-1, whose activation increased the expression of SREBP-1 (Jeon and Osborne 2012). Recent studies showed that PI3K/AKT signaling is significantly upregulated in patients with nonalcoholic fatty liver disease, one of the risk factors for AS, and inhibitors against PI3K and AKT have potential regulatory effects on lipid metabolism (Aljabban et al. 2022). Our results showed that phosphorylation of the PI3K/AKT signaling pathway in the liver of AS model mice is significantly activated, resulting in elevated SREBPs. After 8 weeks of AOB administration, phosphorylation of the PI3K/AKT signaling pathway in the liver is restored, further lowering SREBP-1 signaling in the liver instead of SREBP-2. These results indicate that AOB regulated the PI3K/AKT/SERBP-1 pathway leading to therapeutic actions in AS by reducing lipid levels.

Conclusions

AOB has the therapeutic response of AS, which requires suppression of the PI3K/AKT/SERBP-1 pathway. These were our first findings on AOB’s treatment of AS and the underlying mechanism were associated with inhibition of the PI3K/AKT/SERBP-1 pathway, which suggests that traditional Chinese medicine has an obvious curative effect in the treatment of AS, and had a similar molecular mechanism as other western medicines (Mahtta et al. 2022), which provides strong evidence for our later development and extensive use of traditional Chinese medicine.

Funding Statement

The study was supported by the National Natural Science Foundation of China [82074292] , Jiangsu Traditional Chinese Medicine Science and Technology Development Plan (No. QN202202) Funded by Yushan scholarly sect Classic Traditional Chinese Medicine Classic Inheritance Award Fund.

Consent form

All authors have approved the manuscript and agree with its submission.

Author contributions

Ruiyi Liu, Yan Sun, Boran Zhu and Haoxin Wu designed the study and wrote the manuscript. Ruiyi Liu and Yan Sun performed network pharmacology. Ruiyi Liu, Dong Di and Yan Sun performed the experiments. Ruiyi Liu, Boran Zhu and Yan Sun performed the analyzed the data. All data were generated inhouse, and no paper mill was used. All authors agree to be accountable for all aspects of work ensuring integrity and accuracy.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

The raw data supporting the conclusions of this manuscript will be available from the corresponding author on reasonable request.

References

  1. Aljabban J, Rohr M, Syed S, Khorfan K, Borkowski V, Aljabban H, Segal M, Mukhtar M, Mohammed M, Panahiazar M, et al. 2022. Transcriptome changes in stages of non-alcoholic fatty liver disease. World J Hepatol. 14(7):1382–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aryal A, Harmon AC, Dugas TR.. 2021. Particulate matter air pollutants and cardiovascular disease: strategies for intervention. Pharmacol Ther. 223:107890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Barboza LN, Lívero FA, Prando TB, Ribeiro Rde C, Lourenço EL, Budel JM, de Souza LM, Acco A, Dalsenter PR, Gasparotto AJ.. 2016. Atheroprotective effects of Cuphea carthagenensis (Jacq.) J. F. Macbr. in New Zealand rabbits fed with cholesterol-rich diet. J Ethnopharmacol. 187:134–145. [DOI] [PubMed] [Google Scholar]
  4. Chen Y, Yang W, Guo L, Wu X, Zhang T, Liu J, Zhang J.. 2017. Atractylodes lactone compounds inhibit platelet activation. Platelets. 28(2):194–202. [DOI] [PubMed] [Google Scholar]
  5. Francis GA. 2010. The complexity of HDL. Biochim Biophys Acta. 1801(12):1286–1293. [DOI] [PubMed] [Google Scholar]
  6. Ge L, Liu D, Mao X, Liu S, Guo J, Hou L, Chen X, Huang K.. 2022. Low dose of deoxynivalenol aggravates intestinal inflammation and barrier dysfunction induced by enterotoxigenic Escherichia coli infection through activating macroautophagy/NLRP3 inflammasomes. J Agric Food Chem. 70(9):3009–3022. [DOI] [PubMed] [Google Scholar]
  7. Geovanini GR, Libby P.. 2018. Atherosclerosis and inflammation: overview and updates. Clin Sci. 132(12):1243–1252. [DOI] [PubMed] [Google Scholar]
  8. Hamosh A, Scott AF, Amberger JS, Bocchini CA, McKusick VA.. 2005. Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders. Nucleic Acids Res. 33(Database issue):D514–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Hopkins AL. 2008. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 4(11):682–690. [DOI] [PubMed] [Google Scholar]
  10. Huang H, He Y, Li Y, Gu M, Wu M, Ji L.. 2022. Eriodictyol suppresses the malignant progression of colorectal cancer by downregulating tissue specific transplantation antigen P35B (TSTA3) expression to restrain fucosylation. Bioengineered. 13(3):5551–5563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Jaquish CE, Mahaney MC, Blangero J, Haffner SM, Stern MP, MacCluer JW.. 1996. Genetic correlations between lipoprotein phenotypes and indicators of sex hormone levels in Mexican Americans. Atherosclerosis. 122(1):117–125. [DOI] [PubMed] [Google Scholar]
  12. Jeon TI, Osborne TF.. 2012. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol Metab. 23(2):65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Jia CY, Li JY, Hao GF, Yang GF.. 2020. A drug-likeness toolbox facilitates ADMET study in drug discovery. Drug Discov Today. 25(1):248–258. [DOI] [PubMed] [Google Scholar]
  14. Jin HG, Jin Q, Ryun Kim A, Choi H, Lee JH, Kim YS, Lee DG, Woo ER.. 2012. A new triterpenoid from Alisma orientale and their antibacterial effect. Arch Pharm Res. 35(11):1919–1926. [DOI] [PubMed] [Google Scholar]
  15. Karasawa T, Takahashi A, Saito R, Sekiya M, Igarashi M, Iwasaki H, Miyahara S, Koyasu S, Nakagawa Y, Ishii K, et al. 2011. Sterol regulatory element-binding protein-1 determines plasma remnant lipoproteins and accelerates atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 31(8):1788–1795. [DOI] [PubMed] [Google Scholar]
  16. Keir HR, Chalmers JD.. 2022. Neutrophil extracellular traps in chronic lung disease: implications for pathogenesis and therapy. Eur Respir Rev. 31:210241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Kimura I, Kagawa S, Tsuneki H, Tanaka K, Nagashima F.. 2022. Multitasking bamboo leaf-derived compounds in prevention of infectious, inflammatory, atherosclerotic, metabolic, and neuropsychiatric diseases. Pharmacol Ther. 235:108159. [DOI] [PubMed] [Google Scholar]
  18. Kobiyama K, Ley K.. 2018. Atherosclerosis. Circ Res. 123(10):1118–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Koeth RA, Lam-Galvez BR, Kirsop J, Wang Z, Levison BS, Gu X, Copeland MF, Bartlett D, Cody DB, Dai HJ, et al. 2019. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest. 129(1):373–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Li W, Zhi W, Liu F, Zhao J, Yao Q, Niu X.. 2018. Paeoniflorin inhibits VSMCs proliferation and migration by arresting cell cycle and activating HO-1 through MAPKs and NF-κB pathway. Int Immunopharmacol. 54:103–111. [DOI] [PubMed] [Google Scholar]
  21. Lin YP, Fu SN, Li XP, Wang MM, Fang QL, Qiao X, Yan Q, Hua Y.. 2022. Two novel flavonoids with lipid-lowering activity from Yi medicine Shekaqi. Chem Biodivers. 19:e202200363. [DOI] [PubMed] [Google Scholar]
  22. Liu J, Zhang W, Li Y, Li X, Li Y, Guo F.. 2022. Flavonoids extract from the seeds of Psoralea corylifolia L. (PFE) alleviates atherosclerosis in high-fat diet-induced LDLR(−/−) mice. Phytomedicine. 98:153983. [DOI] [PubMed] [Google Scholar]
  23. Mahtta D, Lee MT, Ramsey DJ, Akeroyd JM, Krittanawong C, Khan SU, Sinh P, Alam M, Garratt KN, Schofield RS, et al. 2022. Significant facility-level variation in utilization of and adherence with secondary prevention therapies among patients with premature atherosclerotic cardiovascular disease: insights from the VITAL (Veterans wIth premaTure AtheroscLerosis) Registry7. Cardiovasc Drugs Ther. 36(1):93–102. [DOI] [PubMed] [Google Scholar]
  24. Moslehi A, Hamidi-Zad Z.. 2018. Role of SREBPs in liver diseases: a mini-review. J Clin Transl Hepatol. 6(3):332–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Pan X, Wan R, Wang Y, Liu S, He Y, Deng B, Luo S, Chen Y, Wen L, Hong T, et al. 2022. Salvianolic acid B inhibiting chemically and mechanically activated Piezo1 channels as a mechanism for ameliorating atherosclerosis. Br J Pharmacol. 179(14):3778–3814. [DOI] [PubMed] [Google Scholar]
  26. Pérez-Belmonte LM, Moreno-Santos I, Cabrera-Bueno F, Sánchez-Espín G, Castellano D, Such M, Crespo-Leiro MG, Carrasco-Chinchilla F, Alonso-Pulpón L, López-Garrido M, et al. 2017. Expression of sterol regulatory element-binding proteins in epicardial adipose tissue in patients with coronary artery disease and diabetes mellitus: preliminary study. Int J Med Sci. 14(3):268–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Rebhan M, Chalifa-Caspi V, Prilusky J, Lancet D.. 1997. GeneCards: integrating information about genes, proteins and diseases. Trends Genet. 13(4):163. [DOI] [PubMed] [Google Scholar]
  28. Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, Barengo NC, Beaton AZ, Benjamin EJ, Benziger CP, et al. 2020. Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study. J Am Coll Cardiol. 76(25):2982–3021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Ru J, Li P, Wang J, Zhou W, Li B, Huang C, Li P, Guo Z, Tao W, Yang Y, et al. 2014. TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. J Cheminform. 6(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T.. 2003. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13(11):2498–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Solanki A, Bhatt LK, Johnston TP.. 2018. Evolving targets for the treatment of atherosclerosis. Pharmacol Ther. 187:1–12. [DOI] [PubMed] [Google Scholar]
  32. Soppert J, Lehrke M, Marx N, Jankowski J, Noels H.. 2020. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev. 159:4–33. [DOI] [PubMed] [Google Scholar]
  33. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al. 2017. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45(D1):D362–D368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Xu X, Zhang W, Huang C, Li Y, Yu H, Wang Y, Duan J, Ling Y.. 2012. A novel chemometric method for the prediction of human oral bioavailability. Int J Mol Sci. 13(6):6964–6982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Xu YH, Zhao LJ, Li Y.. 2009. Alisol B acetate induces apoptosis of SGC7901 cells via mitochondrial and phosphatidylinositol 3-kinases/Akt signaling pathways. World J Gastroenterol. 15(23):2870–2877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Zeng Q, Li L, Siu W, Jin Y, Cao M, Li W, Chen J, Cong W, Ma M, Chen K, et al. 2019. A combined molecular biology and network pharmacology approach to investigate the multi-target mechanisms of Chaihu Shugan San on Alzheimer’s disease. Biomed Pharmacother. 120:109370. [DOI] [PubMed] [Google Scholar]
  37. Zhang R, Zhu X, Bai H, Ning K.. 2019. Network pharmacology databases for traditional Chinese medicine: review and assessment. Front Pharmacol. 10:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, Chanda SK.. 2019. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 10(1):1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Zhu C, Cai T, Jin Y, Chen J, Liu G, Xu N, Shen R, Chen Y, Han L, Wang S, et al. 2020. Artificial intelligence and network pharmacology based investigation of pharmacological mechanism and substance basis of Xiaokewan in treating diabetes. Pharmacol Res. 159:104935. [DOI] [PubMed] [Google Scholar]
  40. Zhu B, Zhai Y, Ji M, Wei Y, Wu J, Xue W, Tao WW, Wu H.. 2020. Alisma orientalis beverage treats atherosclerosis by regulating gut microbiota in ApoE−/− mice. Front Pharmacol. 11:570555. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

The raw data supporting the conclusions of this manuscript will be available from the corresponding author on reasonable request.


Articles from Pharmaceutical Biology are provided here courtesy of Taylor & Francis

RESOURCES