CC BY-NC-ND 4.0 · Planta Med 2023; 89(07): 764-772
DOI: 10.1055/a-2058-1199
Natural Product Chemistry & Analytical Studies
Original Papers

LC-MS Analysis of Ginsenosides in Different Parts of Panax quinquefolius and Their Potential for Coronary Disease Improvement

1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Can Kong
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Hairong Hou
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Hongxia Yu
3   Wendeng Daodishen Industry Co. Ltd., Weihai, China
,
Lizhen Wang
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Peihai Li
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Xiaobin Li
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Yun Zhang
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
,
Liwen Han
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
2   College of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
,
Kechun Liu
1   Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
› Author Affiliations
 

Abstract

Seven main ginsenosides, including ginsenoside Re, ginsenoside Rb1, pseudoginsenoside F11, ginsenoside Rb2, ginsenoside Rb3, ginsenoside Rd, and ginsenoside F2, were identified by LC-QTOF MS/MS from root, leaf and flower extracts of Panax quinquefolius. These extracts promoted intersegmental vessel growth in a zebrafish model, indicating their potential cardiovascular health benefits. Network pharmacology analysis was then conducted to reveal the potential mechanisms of ginsenoside activity in the treatment of coronary artery disease. GO and KEGG enrichment analyses elucidated that G protein-coupled receptors played a critical role in VEGF-mediated signal transduction and that the molecular pathways associated with ginsenoside activity are involved in neuroactive ligand–receptor interaction, cholesterol metabolism, the cGMP–PKG signaling pathway, etc. Moreover, VEGF, FGF2, and STAT3 were confirmed as the major targets inducing proliferation of endothelial cells and driving the pro-angiogenic process. Overall, ginsenosides could be potent nutraceutical agents that act to reduce the risks of cardiovascular disease. Our findings will provide a basis to utilize the whole P. quinquefolius plant in drugs and functional foods.


#

Introduction

The herbs Panax quinquefolius, Panax ginseng, and Panax notoginseng, which belong to the Panax genus (Araliaceae), are recognized as important natural sources of active ingredients for promoting health [1], [2]. Panax quinquefolium Linn. is a highly valued herbal remedy that is commonly used in drugs, dietary supplements, and food products. There has been a large demand for its root material in the Asian market. More than 95% of its wild types are consumed in mainland China and adjacent regions [3]. Historically, the plant was native to the USA and Canada. Since Wendeng (China) has a comparable latitude and similar climate to the producing areas in the USA, P. quinquefolius has been cultivated in the county in recent decades [4]. The active components of P. quinquefolius are known as ginsenosides. Previous studies have compared the chemical properties of P. quinquefolius and its congeneric species to unveil the differences in their ginsenoside compositions to enable better quality control [5], [6], [7], [8]. Based on their skeleton, ginsenosides can mainly be divided into the protopanaxadiol types, protopanaxatriol types, oleanolic acid types, etc., which exhibit diverse pharmacological activities such as anti-tumor, cardiovascular protective, immunomodulatory, anti-inflammatory, antidiabetic, and hepatoprotective effects, as well as being useful tonics [9], [10], [11], [12].

The leaves, stems and flower buds of P. quinquefolius contain many of the same active ingredients and have received much attention [13], [14]. Twenty marker ginsenosides have been characterized via untargeted metabolomics analysis as the most important diagnostic markers for differentiating the stem, leaf, flower bud, and root parts [15]. However, the components and bioactivity of the whole plants have not been investigated comprehensively. “Total ginsenosides” from P. quinquefolius stems and leaves have been added to the China Food Drug Administration standards (YBZ01382003). Systematic evaluation of the leaves, stems, and flower buds from P. quinquefolius will contribute to exploring the therapeutic basis of these plants and elaborating on the quality standards to promote their further utilization. In our study, liquid chromatography-quadrupole-time of flight mass spectrometry (LC-Q-TOF-MS) was applied for the analysis of ginsenosides in different parts (root, leaves, and flower buds) of P. quinquefolius. The compounds were characterized by accurate mass measurements, fragment ions, and comparing retention times to the reference standards. The relationships between the components and their cardio-protective effects were established via an in vivo zebrafish model, network pharmacology, and investigating the molecular mechanism in human umbilical vein endothelial cells (HUVECs). Our work lays a foundation for the utilization of the species in health-promoting products.


#

Results

The total ion chromatograms of root, leaf, and flower extracts (RE, LE, and FE) from P. quinquefolius are shown in [Fig. 1], and seven ginsenosides were successfully identified by LC-MS/MS under Q/TOF conditions through a comparison with data from the literature [16], [17], [18], [19]. In negative-ion mode, the MS/MS data of these ginsenosides were always acquired from the adduct ions, which provided valuable structural information. Ginsenoside Re (1) showed a [M + Cl] ion at m/z 981.5102 and characteristic fragment ions at m/z 945 [M – H], 783, 647, and 475. Ginsenoside Rb1 (2) generated a [M + Cl] ion at m/z 1143.5580 and corresponding fragment ions at m/z 1107 [M – H], 945, 783 and 621. Pseudoginsenoside F11 (3) exhibited a [M + Cl] quasi-molecular ion peak at m/z 835.4566 with daughter ions at m/z 799 [M – H], 653, and 491. Ginsenoside Rb2 (4) displayed a [M + Cl] ion at m/z 1113.5393 and fragment ions at m/z 1077 [M – H], 945, 783, and 621. Ginsenoside Rb3 (5) produced a [M + Cl] ion at m/z 1113.5513 along with fragment ions at m/z 1077 [M – H], 945, 783, and 621. Ginsenoside Rd (6) showed a [M + Cl] ion at m/z 981.5117 and main fragment ions at m/z 945 [M – H], 783, and 621. Ginsenoside F2 (7) demonstrated a signal corresponding to the [M + Cl] ion at m/z 819.4597 and fragment ions at m/z 621. Lastly, all of the compounds were confirmed by LC–MS analysis of the reference substances (Fig. 1S–7S, Supporting Information). The ginsenosides Re and Rb1 were detected at high levels in the roots, and the ginsenosides Re, Rb2, Rb3, Rd, F2, and pseudoginsenoside F11 were abundant in the leaves and flowers. The details and structures of the identified ginsenosides are summarized in Table 1S and Fig. 8S (Supporting Information), respectively.

Zoom Image
Fig. 1 LC-MS chromatograms of leaf (a), flower (b) and root (c) extracts from P. quinquefolius. The peaks were identified as ginsenoside Re (1), ginsenoside Rb1 (2), pseudoginsenoside F11 (3), ginsenoside Rb2 (4), ginsenoside Rb3 (5), ginsenoside Rd (6), and ginsenoside F2 (7).

Coronary artery disease (CAD) is among the cardiovascular disease entities and will soon be the leading cause of death globally. Therapeutic angiogenesis is a promising strategy for revolutionizing the treatment of CAD, and the components for stimulating the growth of new blood vessels in the heart are highlighted in current clinical trials [20], [21]. Many proteins necessary for blood vessel growth in zebrafish are highly conserved and the same as those in mammals. Therefore, a transgenic zebrafish (Tg : vegfr2-GFP) model containing fluorescent blood vessels was considered to be an ideal tool for evaluating the effect of pro-angiogenesis compounds [22]. The zebrafish larvae had well-developed intersegmental blood vessels (ISVs) in the control, which were connected to the dorsal longitudinal anastomotic vessels. In contrast to the intact interstitial vessels, the vascular morphology of larvae showed severe damage after the PTK787 treatment, resulting in an obvious impairment of ISV formation in zebrafish. RE, LE, and FE were applied to determine whether angiogenesis could resume with the increase in treatment concentrations. After application, the extracts were shown to decrease the proportion of defective blood vessels and increase the proportion of normal blood vessels at proper concentrations. In particular, angiogenesis was more pronounced under 10 µg/mL LE, 25 µg/mL RE, and 10 µg/mL–25 µg/mL FE treatments, and these were statistically significant for restoring PTK787-induced ISV insufficiency (Table 2S, Supporting Information). The results indicated that RE, LE, and FE have the potential to promote angiogenesis; however, interestingly, the extracts exerted an inverted effect on protective vessels at concentrations greater than 50 µg/mL. Danhong injection (DHI), a Chinese patent compound injection, was used as the positive control.

Network pharmacology has been proven to be a dominant paradigm by establishing a visualization network to understand the complex pharmacological action of effective substances in herbs [23]. After creating the intersections via an online Venn diagram (http://bioinformatics.psb.ugent.be/webtools/Venn/), 24 targets of the seven ginsenosides and 19 targets for CAD were gathered, respectively, to predict the links between the compounds and diseases through protein–protein interaction (PPI). “Degree” is the number of edges that a node shares with others, which is used to estimate the importance of the node in complex networks. As shown in [Fig. 3], the analysis of the network revealed that VEGF (Degree, 22), FGF2 (Degree, 18) and STAT3 (Degree, 20) were visualized as the major targets of the compounds. Ginsenoside F2 and pseudoginsenoside F11 were selected as the core compounds. Remarkably, both VEGF and FGF2 act, as angiogenesis factors can be significantly affected by the two compounds, triggering the STAT3 targets and then hitting downstream genes for the treatment of CAD. The general network properties of the targets are shown in Table 3S (Supporting Information).

Zoom Image
Fig. 2 Pro-angiogenic effects of RE, LE, and FE in transgenic zebrafish (Tg : vegfr2-GFP). The intersegmental vessels are indicated by the white square; Scale bar, 200 µm.
Zoom Image
Fig. 3 Construction of a candidate-target network for ginsenosides against CAD. The triangle nodes represent compounds; the circular nodes and rectangle nodes represent predicted targets and therapeutic targets, respectively.

GO enrichment analysis was performed with the whole human genome as the background. The GO biological process and GO molecular function were used to classify the genes according to their functional annotation ([Fig. 4]). Among biological processes, the candidate targets were mainly related to vascular processes in the circulatory system (P = 1.51 × 10−15), including regulation of blood vessel size, regulation of blood vessel diameter, vasoconstriction, etc. Among molecular functions, the targets were enriched in G protein-coupled receptors (GPCRs) activity, alpha2-adrenergic receptor activity, serotonin receptor activity, intermembrane lipid transfer activity, transmembrane signaling receptor activity, etc. GPCRs are proven to produce the most significant P-values, and indeed a number of discussions have shown that they play a critical role in VEGF-mediated signal transduction and are associated with the field of angiogenesis [24]. The KEGG pathway enrichment analysis was carried out to elucidate the molecular pathways involving these targets. The top 20 pathways are shown as the core pathways in [Fig. 5]. The results indicated that the KEGG pathways of the ginsenosides against CAD include neuroactive ligand–receptor interaction (10 genes), cholesterol metabolism (8 genes), the cGMP-PKG signaling pathway (6 genes), etc. Neuroactive ligand–receptor interactions have been suggested to be an important factor in response to angiogenesis, yielding a P-value of 1.55 × 10−6 among these terms. An explanation was that the growth of blood vessels and nerves promote each other and follow the same pathway [25]. Furthermore, we performed molecular biological assays involving VEGF, FGF2, and STAT3 to validate the effects of the compounds.

Zoom Image
Fig. 4 GO enrichment analysis displayed the targets from PPI network are enriched in various biological processes and molecular function.
Zoom Image
Fig. 5 KEGG analysis revealed the top 20 pathway terms associated with the targets from PPI network.

Because of their importance in angiogenesis, ginsenoside F2 (protopanaxadiol-type) and pseudoginsenoside F11 (ocotillol-type) were investigated as the representative compounds for their effects on vascular growth. According to the protocol, the molecular mechanisms involved in the angiogenic process were generalized to HUVECs [26], [27]. In [Fig. 6 a] – [b], we observed that the exposure of HUVECs to ginsenoside F2 and pseudoginsenoside F11 resulted in an obvious increase in cell numbers at concentrations of 2.5 – 5 µM and 5 – 10 µM, respectively, which exhibit statistically significant differences with P-values < 0.01. Cells treated with 20 ng/mL VEGF were used as the positive control. The results supported that HUVEC proliferation was allowed to proceed in the presence of the compounds at individual-level concentrations, whereas endothelial cell proliferation has been recognized as the key steps of the angiogenic process. However, similar to the zebrafish model, ginsenoside F2 and pseudoginsenoside F11 also exerted an inverted effect on cell viability at higher concentrations (more than 10 µM and 25 µM, respectively). As featured in [Fig. 6 c] – [d], the compounds can activate expression of the proteins as predicted by bioinformatics. When compared with the control, the Western blotting data produced a dose-dependent increase in the VEGF, FGF2, and p-STAT3 levels at concentrations of 1 – 5 µM ginsenoside F2 and 2.5 – 10 µM pseudoginsenoside F11, respectively. Although the expression of p-STAT3 protein was obviously enhanced, the total level of STAT3 was not affected. Consequently, VEGF, FGF2, and p-STAT3 were recognized as the potential influencers of the ginsenosides, stimulating the proliferation of endothelial cells and driving the pro-angiogenic process.

Zoom Image
Fig. 6 The ginsenosides stimulated the proliferation and induced VEGF, FGF2, and p-STAT3 activation in HUVECs. (A) Proliferation effects of ginsenoside F2, (B) Proliferation effects of pseudoginsenoside F11, (C) Ginsenoside F2-mediated alterations in protein expression, (D) Pseudoginsenoside F11-mediated alterations in protein expression. (The experiments were repeated six and three times for cell viability and western blot, respectively; error bars represent means ± SD, *P < 0.05 and ** P < 0.01 vs. control.)

#

Discussion

Ginsenosides accumulate at high levels in many parts of P. quinquefolius, such as the roots, leaves, and flower buds. In this study, the ginsenosides Re and Rb1 were detected at high levels in the roots. Ginsenosides Rb1 has been recognized as the top marker in the roots, which must meet the essential requirements for medical use in the Chinese Pharmacopoeia. Previous work showed that the leaves and flowers exhibited similar ginsenoside compositions, and pseudoginsenoside F11 was found to be the characteristic component in P quinquefolium [15]. Consistent with the conclusion, the protopanaxadiol-type ginsenosides Rb2, Rb3, Rd, F2, protopanaxatriol-type ginsenosides Re, and ocotillol-type pseudoginsenoside F11 were identified from total-ion chromatograms of the leaves and flowers with high intensity. Because of the cultivation period of 4 – 6 years for the roots, the leaves and flowers can be harvested every year, and they are an important resource for obtaining these compounds.

There is evidence that P. quinquefolius can offer benefit to patients with heart failure and has been purported to improve cardiac performance. The ginsenosides were demonstrated to be the active constituents that protect against myocardial infarction and thereby ameliorate cardiac dysfunction [28], [29]. The present study showed that RE, LE, and FE can restore vascular insufficiency in zebrafish at the specified concentrations and verified their positive association with angiogenic factors, illustrating their potential protective effects toward cardiovascular diseases. Neuroactive ligand–receptor interaction, cholesterol metabolism, and the cGMP–PKG signaling pathway were indicated as the top KEGG enrichments for the ginsenosides against CAD. Accumulating clinical evidence has confirmed that these pathways play a crucial role in angiogenesis, vascular protection, and vasodilatory effects [30], [31], [32].

Angiogenesis involves endothelial cell differentiation, proliferation, migration, and cord formation for the formation of new capillaries sprouting from pre-existing vessels [33]. Many endothelium-specific molecules are associated with this process. For example, VEGF is present and is a key driver of angiogenesis signaling pathways. Recent evidence has demonstrated that VEGF significantly induces STAT3 activation, which is essential for vascular endothelial cell proliferation, vascular survival, or remodeling. Interestingly, STAT3 is also phosphorylated in response to FGF2 during angiogenic activation [34], [35], [36]. Based on our studies, we suggested that the overexpression of VEGF and FGF2 was induced by the ginsenosides and triggered the phosphorylation of STAT3. They were responsible for the proliferation of endothelial cells, leading to the angiogenic response. Our findings will provide a basis to facilitate the utilization of P. quinquefolius in nutraceutical agents and functional foods for CAD treatment.

We analyzed molecular signals at concentrations within the range shown to have a promoting effect on HUVEC proliferation. However, as previously reported, the ginsenosides were also found to inhibit the proliferation of HUVECs and served as the inhibitors against vascular growth at higher levels, while also being possibly involved in the expression of various factors associated with angiogenesis via the key mediator of VEGF. These qualities make P. quinquefolius potentially a very promising agent for controlling tumor growth [37], [38]. To better evaluate the biological characteristics and effects, the mechanisms involved in the cardiovascular activities of these ginsenosides should be further investigated.


#

Materials and Methods

Chemicals and reagents

Ginsenoside Re, ginsenoside Rb1, pseudoginsenoside F11, ginsenoside Rb2, ginsenoside Rb3, ginsenoside Rd, and ginsenoside F2 were purchased from Shanghai Yuanye Biotechnology Co., Ltd. MS-grade water and acetonitrile were acquired from Watsons Ltd and Tedia Company Inc., respectively. All other chemicals used were analytical grade. Danhong injection (DHI, lot: 16011017) was purchased from Danhong Pharmaceutical Co., Ltd. Human umbilical vein endothelial cells (HUVEC) were acquired from the American Type Culture Collection (ATCC).


#

Plant material extraction

Root, leaves, and flower buds of P. quinquefolius were obtained from Wendeng Daodishen Industry Co., Ltd. The samples were identified by Professor Kechun Liu, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences). The plant materials were deposited at the Key Laboratory for Drug Screening Technology of the Biology Institute as a voucher specimen (No. SWS402B). One gram each of powdered root, leaf, and flower material was extracted at 50 °C with 8 mL of 50% ethanol for 1 h using ultrasound.


#

LC–MS/MS analysis

The extract solutions were filtered through 0.45 µm nylon filters and subjected to LC-Q/TOF-MS analysis. LC-MS was performed on an XDB-C18 HPLC column (4.6 × 250 mm, 5 µm; Agilent), and the gradient conditions for solvents A (water) and B (acetonitrile) were as follows: 2 – 35% B (0 – 15 min), 35% B (15 – 25 min), 35 – 60% B (25 – 40 min), and 60 – 90% B (40 – 45 min). The following MS conditions were employed: ESI-negative-ion mode, nebulizer pressure at 35 psi, drying gas temperature of 325 °C, drying gas flow of 10 L/min, capillary voltage of 4000 V, and scanning range of 200 – 2000 m/z. High-resolution tandem mass spectrometry (HR–MS/MS) was performed for qualitative identification using standard compounds as a reference.


#

Zebrafish angiogenesis assay

Transgenic zebrafish (Tg : vegfr2-GFP) were obtained from the Zebrafish Drug Screening Platform, Qilu University of Technology (Shandong Academy of Sciences). After removal of the solvent, the residues of root, leaf, and flower bud extracts were evaluated for their angiogenesis activity. Zebrafish larvae 24 hpf (hour post-fertilization) were divided randomly into 24-well plates at a density of 10 larvae per well. The experiment consisted of a vehicle control group (embryo medium), a model group (0.1 µg/mL PTK787), a positive group (0.1 µg/mL PTK787 + 10 µL/mL DHI), and intervention groups (0.1 µg/mL PTK787 + 10, 25, 50, 100, or 150 µg/mL of each extract). All treatments, performed in triplicate, were maintained under standard culture conditions for a further 24 h. Subsequently, the zebrafish larvae were observed under a fluorescence microscope (SZX16, Olympus), and the angiogenic activity was assessed according to the integrity of the intersegmental vessels.


#

Target database construction and bioinformatics analysis

The chemical structures of ginsenoside were downloaded from the PubChem database (http://pubchem.ncbi.nlm.nih.gov/) and used for network pharmacology analysis. Swiss Target Prediction (http://www.swisstargetprediction.ch/) was used to screen the potential target of the active components. Considering the evidence for angiogenesis, the target genes associated with “Coronary artery disease” (CAD) were collected using the DisGeNET (http://www.disgenet.org/) and GeneCards (https://www.genecards.org/) databases. The candidate targets were inputted to String 11.5 (https://string-db.org/) to obtain the relevant information on protein interactions [39]. Furthermore, a compound-target network was constructed by employing Cytoscape 3.6.1. to study the therapeutic mechanism of CAD. GO and KEGG pathway enrichment analyses were executed using the OmicShare tools (https://www.omicshare.com/tools).


#

HUVEC proliferation and Western blot assay

The cells were cultured in RPMI-1640 medium under 5% CO2 at 37 °C. An MTT assay was used to evaluate the effects of pseudoginsenoside F11 and ginsenoside F2 on cell proliferation, which is responsible for angiogenesis. Furthermore, Western blot was performed as previously described with minor modifications [40]. Briefly, after lysing cells in RIPA buffer, the proteins were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently transferred to polyvinylidene fluoride (PVDF) membranes. The primary antibodies and horseradish peroxidase-conjugated secondary antibodies were incubated with the membranes, and the protein bands were visualized using an enhanced chemiluminescence (ECL) substrate (cat No. 180 – 5001, Tanon) with the Tanon 5200 system.

The following antibodies were used: anti-FGF2 (1 : 1000, cat No. PA5-116495, Invitrogen), anti-STAT3 (1 : 1000, cat No. 60199-1-Ig, Proteintech), anti-p-STAT3 (1 : 1000, cat No. E121 – 31, abcam), anti-VEGF (1 : 1000, cat No. 66 828-1-Ig, Proteintech), anti-β-actin (1 : 1000, cat No. 200 068 – 8F10, ZenBio), HRP-conjugated goat anti-rabbit IgG (1 : 5000, cat No. A0208, Beyotime), and HRP-conjugated goat anti-mouse IgG (1 : 5000, cat No. A0216, Beyotime).


#

Statistics and analysis

In biological assays for multiple comparisons, ANOVA tests were performed using an online tool, Variance Calculator 20210415 (AB126 Software Park, http://www.ab126.com/shuxue/8016.html). Differences with a P value of < 0.05 were considered to be statistically significant.


#
#

Contributorsʼ Statement

Conception and design of the work: X. Zhang, L. Han, K. Liu; data collection: X. Zhang, C. Kong, X. Wang, H. Hou, H. Yu; analysis and interpretation of the data: X. Zhang, X. Wang, L. Wang, P. Li, X. Li, Y. Zhang; statistical analysis: X. Zhang, C. Kong; drafting the manuscript: X. Zhang, X Wang, L. Wang, X. Li, Y. Zhang; critical revision of the manuscript: X. Zhang, L. Han, K. Liu.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Funding

The study was financially supported by the Major Scientific and Technological Innovation Projects of Shandong Province (grant number: 2021CXGC010507), the Taishan Scholar Project from Shandong Province to H.L (grant number: ts20190950), and the Jinan Talent Project for University (grant number: 2021GXRC047).

Supporting Information

  • References

  • 1 Jee HS, Chang KH, Park SH, Kim KT, Paik HD. Morphological characterization, chemical components, and biofunctional activities of Panax ginseng, Panax quinquefolium, and Panax notoginseng roots: A comparative study. Food Reviews International 2014; 30: 91-111
  • 2 Kim DH. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng . J Ginseng Res 2012; 36: 1-15
  • 3 Liu H, Burkhart EP, Chen VYJ, Wei X. Promotion of in situ forest farmed American ginseng (Panax quinquefolius L.) as a sustainable use strategy: Opportunities and challenges. Frontiers in Ecology and Evolution 2021; 9: 652103
  • 4 Zhang XM, Lu XH, Jiao XL, Bi YM, Gao WW. First report of Ilyonectria vredehoekensis causing rusty root on American ginseng in China. Plant Dis 2019; 103: 2944
  • 5 Du ZX, Li JH, Zhang X, Pei J, Huang LF. An integrated LC-MS-based strategy for the quality assessment and discrimination of three Panax species. Molecules 2018; 23: 2988
  • 6 Yang WZ, Qiao X, Li K, Fan JR, Bo T, Guo DA, Ye M. Identification and differentiation of Panax ginseng, Panax quinquefolium, and Panax notoginseng by monitoring multiple diagnostic chemical markers. Acta Pharm Sin B 2016; 6: 568-575
  • 7 Yang WZ, Shi XJ, Yao CL, Huang Y, Hou JJ, Han SM, Feng ZJ, Wei WL, Wu WY, Guo DA. A novel neutral loss/product ion scan-incorporated integral approach for the untargeted characterization and comparison of the carboxyl-free ginsenosides from Panax ginseng, Panax quinquefolius, and Panax notoginseng . J Pharm Biomed Anal 2020; 177: 112813
  • 8 Shi XJ, Yang WZ, Qiu S, Yao CL, Shen Y, Pan HQ, Bi QR, Yang M, Wu WY, Guo DA. An in-source multiple collision-neutral loss filtering based nontargeted metabolomics approach for the comprehensive analysis of malonyl-ginsenosides from Panax ginseng, P. quinquefolius, and P. notoginseng . Anal Chim Acta 2017; 952: 59-70
  • 9 Hou MQ, Wang RF, Zhao SJ, Wang ZT. Ginsenosides in Panax genus and their biosynthesis. Acta Pharm Sin B 2021; 11: 1813-1834
  • 10 Mancuso C, Santangelo R. Panax ginseng and Panax quinquefolius: From pharmacology to toxicology. Food Chem Toxicol 2017; 107: 362-372,
  • 11 Yuan CS, Wang CZ, Wicks SM, Qi LW. Chemical and pharmacological studies of saponins with a focus on American ginseng. J Ginseng Res 2010; 34: 160-167
  • 12 Ratan ZA, Haidere MF, Hong YH, Park SH, Lee JO, Lee JS, Cho JY. Pharmacological potential of ginseng and its major component ginsenosides. J Ginseng Res 2021; 45: 199-210
  • 13 Ma ZN, Li YZ, Li W, Yan XT, Yang G, Zhang J, Zhao LC, Yang LM. Nephroprotective effects of saponins from leaves of Panax quinquefolius against cisplatin-induced acute kidney injury. Int J Mol Sci 2017; 18: 1407
  • 14 Li F, Lv CN, Li Q, Wang J, Song D, Liu PP, Zhang DD, Lu JC. Chemical and bioactive comparison of flowers of Panax ginseng Meyer, Panax quinquefolius L., and Panax notoginseng Burk. J Ginseng Res 2017; 41: 487-495
  • 15 Wang HD, Zhang CX, Zuo TT, Li WW, Jia L, Wang XY, Qian YX, Guo D, Yang WZ. In-depth profiling, characterization, and comparison of the ginsenosides among three different parts (the root, stem leaf, and flower bud) of Panax quinquefolius L. by ultra-high performance liquid chromatography/quadrupole-Orbitrap mass spectrometry. Anal Bioanal Chem 2019; 411: 7817-7829
  • 16 Li SL, Lai SF, Song JZ, Qiao CF, Liu X, Zhou Y, Cai H, Cai BC, Xu HX. Decocting-induced chemical transformations and global quality of Du-Shen-Tang, the decoction of ginseng evaluated by UPLC-Q-TOF-MS/MS based chemical profiling approach. J Pharm Biomed Anal 2010; 53: 946-957
  • 17 Ma XQ, Xiao HB, Liang XM. Identification of ginsenosides in Panax quinquefolium by LC-MS. Chromatographia 2006; 64: 31-36
  • 18 Mao Q, Bai M, Xu JD, Kong M, Zhu LY, Zhu H, Wang Q, Li SL. Discrimination of leaves of Panax ginseng and P. quinquefolius by ultra high performance liquid chromatography quadrupole/time-of-flight mass spectrometry based metabolomics approach. J Pharm Biomed Anal 2014; 97: 129-140
  • 19 Wang J, Liu H, Gao WY, Zhang LM. Comparison of ginsenoside composition in native roots and cultured callus cells of Panax quinquefoliumL . Acta Physiol Plant 2013; 35: 1363-1366
  • 20 Malakar AK, Choudhury D, Halder B, Paul P, Uddin A, Chakraborty S. A review on coronary artery disease, its risk factors, and therapeutics. J Cell Physiol 2019; 234: 16812-16823
  • 21 Ryan CT, Patel V, Rosengart TK. Clinical potential of angiogenic therapy and cellular reprogramming. JTCVS Open 2021; 6: 108-115
  • 22 Zhang XM, Shi YP, Wang LZ, Li XB, Zhang SS, Wang XM, Jin M, Hsiao CD, Lin HW, Han LW, Liu KC. Metabolomics for biomarker discovery in fermented black garlic and potential bioprotective responses against cardiovascular diseases. J Agric Food Chem 2019; 44: 12191-12198
  • 23 Wang X, Wang ZY, Zheng JH, Li S. TCM network pharmacology: A new trend towards combining computational, experimental and clinical approaches. Chin J Nat Med 2021; 19: 1-11
  • 24 Richard DE, Vouret-Craviari V, Pouysségur J. Angiogenesis and G-protein-coupled receptors: Signals that bridge the gap. Oncogene 2001; 20: 1556-1562
  • 25 Zhang XD, Liu LG, Liu DY, Li YT, He J, Shen L. 17β-Estradiol promotes angiogenesis of bone marrow mesenchymal stem cells by upregulating the PI3K-Akt signaling pathway. Comput Struct Biotechnol J 2022; 20: 3864-3873
  • 26 Latifi-Navid H, Soheili ZS, Samiei S, Sadeghi M, Taghizadeh S, Pirmardan ER, Ahmadieh H. Network analysis and the impact of Aflibercept on specific mediators of angiogenesis in HUVEC cells. J Cell Mol Med 2021; 25: 8285-8299
  • 27 Lam HW, Lin HC, Lao SC, Gao JL, Hong SJ, Leong CW, Yue PYK, Kwan YW, Leung AYH, Wang YT, Lee SMY. The angiogenic effects of Angelica sinensis extract on HUVEC in vitro and zebrafish in vivo. J Cell Biochem 2008; 103: 195-211
  • 28 Jiang M, Murias JM, Chrones T, Sims SM, Lui E, Noble EG. American ginseng acutely regulates contractile function of rat heart. Front Pharmacol 2014; 5: 43
  • 29 Kim JH. Pharmacological and medical applications of Panax ginseng and ginsenosides: a review for use in cardiovascular diseases. J Ginseng Res 2018; 42: 264-269
  • 30 Yao QY, Huang YQ, Liu AD, Zhu MZ, Liu J, Yan H, Zhang QY, Geng B, Gao YS, Du SX, Huang P, Tang CS, Du JB, Jin HF. The vasodilatory effect of sulfur dioxide via SGC/cGMP/PKG pathway in association with sulfhydryl-dependent dimerization. Am J Physiol Regul Integr Comp Physiol 2016; 310: R1073-R1080
  • 31 Zhang K, Song W, Li DL, Jin X. Apigenin in the regulation of cholesterol metabolism and protection of blood vessels. Exp Ther Med 2017; 13: 1719-1724
  • 32 Liu J, Wang XD, Lin JH, Li SH, Deng GX, Wei JR. Classifiers for predicting coronary artery disease based on gene expression profiles in peripheral blood mononuclear cells. Int J Gen Med 2021; 14: 5651-5663
  • 33 Yoodee S, Peerapen P, Plumworasawat S, Thongboonkerd V. ARID1A knockdown in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity. Int J Biol Macromol 2021; 180: 1-13
  • 34 Pulkkinen HH, Kiema M, Lappalainen JP, Toropainen A, Beter M, Tirronen A, Holappa L, Niskanen H, Kaikkonen MU, Ylä-Herttuala S, Laakkonen JP. BMP6/TAZ-Hippo signaling modulates angiogenesis and endothelial cell response to VEGF. Angiogenesis 2021; 24: 129-144
  • 35 Chen SH, Murphy D, Lassoued W, Thurston G, Feldman MD, Lee WMF. Activated STAT3 is a mediator and biomarker of VEGF endothelial activation. Cancer Biol Ther 2008; 7: 1994-2003
  • 36 Yang X, Qiao D, Meyer K, Friedl A. Signal transducers and activators of transcription mediate fibroblast growth hactor-induced vascular endothelial morphogenesis. Cancer Res 2009; 69: 1668-1677
  • 37 Nakhjavani M, Smith E, Townsend AR, Price TJ, Hardingham JE. Anti-Angiogenic Properties of Ginsenoside Rg3. Molecules 2020; 25: 4905
  • 38 Yue PYK, Wong DYL, Wu PK, Leung PY, Mak NK, Yeung HW, Liu L, Cai ZW, Jiang ZH, Fan TPD, Wong RNS. The angiosuppressive effects of 20(R)-ginsenoside Rg3. Biochem Pharmacol 2006; 72: 437-445
  • 39 Che YH, Xu ZR, Ni LL, Dong XX, Yang ZZ, Yang ZB. Isolation and identification of the components in Cybister chinensis Motschulsky against inflammation and their mechanisms of action based on network pharmacology and molecular docking. J Ethnopharmacol 2022; 285: 114851
  • 40 Shi L, Yang F, Luo F, Liu Y, Zhang F, Zou MJ, Liu QZ. Evodiamine exerts anti-tumor effects against hepatocellular carcinoma through inhibiting β-catenin-mediated angiogenesis. Tumor Biol 2016; 37: 12791-12803

Correspondence

Professor Liwen Han
Biology Institute
Qilu University of Technology (Shandong Academy of Sciences)
No. 28789 Jingshidong Road, Caishi Town, Licheng District
250103 Jinan
China   
Phone: + 86 05 31 82 60 53 31   
Fax: + 86 05 31 82 60 53 52   

 

Professor Kechun Liu
Biology Institute
Qilu University of Technology (Shandong Academy of Sciences)
No. 28789 Jingshidong Road, Caishi Town, Licheng District
250103 Jinan
China   
Phone: + 86 05 31 82 60 53 31   
Fax: + 86 05 31 82 60 53 52   

Publication History

Received: 26 September 2022

Accepted after revision: 12 March 2023

Accepted Manuscript online:
20 March 2023

Article published online:
17 April 2023

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commecial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Jee HS, Chang KH, Park SH, Kim KT, Paik HD. Morphological characterization, chemical components, and biofunctional activities of Panax ginseng, Panax quinquefolium, and Panax notoginseng roots: A comparative study. Food Reviews International 2014; 30: 91-111
  • 2 Kim DH. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng . J Ginseng Res 2012; 36: 1-15
  • 3 Liu H, Burkhart EP, Chen VYJ, Wei X. Promotion of in situ forest farmed American ginseng (Panax quinquefolius L.) as a sustainable use strategy: Opportunities and challenges. Frontiers in Ecology and Evolution 2021; 9: 652103
  • 4 Zhang XM, Lu XH, Jiao XL, Bi YM, Gao WW. First report of Ilyonectria vredehoekensis causing rusty root on American ginseng in China. Plant Dis 2019; 103: 2944
  • 5 Du ZX, Li JH, Zhang X, Pei J, Huang LF. An integrated LC-MS-based strategy for the quality assessment and discrimination of three Panax species. Molecules 2018; 23: 2988
  • 6 Yang WZ, Qiao X, Li K, Fan JR, Bo T, Guo DA, Ye M. Identification and differentiation of Panax ginseng, Panax quinquefolium, and Panax notoginseng by monitoring multiple diagnostic chemical markers. Acta Pharm Sin B 2016; 6: 568-575
  • 7 Yang WZ, Shi XJ, Yao CL, Huang Y, Hou JJ, Han SM, Feng ZJ, Wei WL, Wu WY, Guo DA. A novel neutral loss/product ion scan-incorporated integral approach for the untargeted characterization and comparison of the carboxyl-free ginsenosides from Panax ginseng, Panax quinquefolius, and Panax notoginseng . J Pharm Biomed Anal 2020; 177: 112813
  • 8 Shi XJ, Yang WZ, Qiu S, Yao CL, Shen Y, Pan HQ, Bi QR, Yang M, Wu WY, Guo DA. An in-source multiple collision-neutral loss filtering based nontargeted metabolomics approach for the comprehensive analysis of malonyl-ginsenosides from Panax ginseng, P. quinquefolius, and P. notoginseng . Anal Chim Acta 2017; 952: 59-70
  • 9 Hou MQ, Wang RF, Zhao SJ, Wang ZT. Ginsenosides in Panax genus and their biosynthesis. Acta Pharm Sin B 2021; 11: 1813-1834
  • 10 Mancuso C, Santangelo R. Panax ginseng and Panax quinquefolius: From pharmacology to toxicology. Food Chem Toxicol 2017; 107: 362-372,
  • 11 Yuan CS, Wang CZ, Wicks SM, Qi LW. Chemical and pharmacological studies of saponins with a focus on American ginseng. J Ginseng Res 2010; 34: 160-167
  • 12 Ratan ZA, Haidere MF, Hong YH, Park SH, Lee JO, Lee JS, Cho JY. Pharmacological potential of ginseng and its major component ginsenosides. J Ginseng Res 2021; 45: 199-210
  • 13 Ma ZN, Li YZ, Li W, Yan XT, Yang G, Zhang J, Zhao LC, Yang LM. Nephroprotective effects of saponins from leaves of Panax quinquefolius against cisplatin-induced acute kidney injury. Int J Mol Sci 2017; 18: 1407
  • 14 Li F, Lv CN, Li Q, Wang J, Song D, Liu PP, Zhang DD, Lu JC. Chemical and bioactive comparison of flowers of Panax ginseng Meyer, Panax quinquefolius L., and Panax notoginseng Burk. J Ginseng Res 2017; 41: 487-495
  • 15 Wang HD, Zhang CX, Zuo TT, Li WW, Jia L, Wang XY, Qian YX, Guo D, Yang WZ. In-depth profiling, characterization, and comparison of the ginsenosides among three different parts (the root, stem leaf, and flower bud) of Panax quinquefolius L. by ultra-high performance liquid chromatography/quadrupole-Orbitrap mass spectrometry. Anal Bioanal Chem 2019; 411: 7817-7829
  • 16 Li SL, Lai SF, Song JZ, Qiao CF, Liu X, Zhou Y, Cai H, Cai BC, Xu HX. Decocting-induced chemical transformations and global quality of Du-Shen-Tang, the decoction of ginseng evaluated by UPLC-Q-TOF-MS/MS based chemical profiling approach. J Pharm Biomed Anal 2010; 53: 946-957
  • 17 Ma XQ, Xiao HB, Liang XM. Identification of ginsenosides in Panax quinquefolium by LC-MS. Chromatographia 2006; 64: 31-36
  • 18 Mao Q, Bai M, Xu JD, Kong M, Zhu LY, Zhu H, Wang Q, Li SL. Discrimination of leaves of Panax ginseng and P. quinquefolius by ultra high performance liquid chromatography quadrupole/time-of-flight mass spectrometry based metabolomics approach. J Pharm Biomed Anal 2014; 97: 129-140
  • 19 Wang J, Liu H, Gao WY, Zhang LM. Comparison of ginsenoside composition in native roots and cultured callus cells of Panax quinquefoliumL . Acta Physiol Plant 2013; 35: 1363-1366
  • 20 Malakar AK, Choudhury D, Halder B, Paul P, Uddin A, Chakraborty S. A review on coronary artery disease, its risk factors, and therapeutics. J Cell Physiol 2019; 234: 16812-16823
  • 21 Ryan CT, Patel V, Rosengart TK. Clinical potential of angiogenic therapy and cellular reprogramming. JTCVS Open 2021; 6: 108-115
  • 22 Zhang XM, Shi YP, Wang LZ, Li XB, Zhang SS, Wang XM, Jin M, Hsiao CD, Lin HW, Han LW, Liu KC. Metabolomics for biomarker discovery in fermented black garlic and potential bioprotective responses against cardiovascular diseases. J Agric Food Chem 2019; 44: 12191-12198
  • 23 Wang X, Wang ZY, Zheng JH, Li S. TCM network pharmacology: A new trend towards combining computational, experimental and clinical approaches. Chin J Nat Med 2021; 19: 1-11
  • 24 Richard DE, Vouret-Craviari V, Pouysségur J. Angiogenesis and G-protein-coupled receptors: Signals that bridge the gap. Oncogene 2001; 20: 1556-1562
  • 25 Zhang XD, Liu LG, Liu DY, Li YT, He J, Shen L. 17β-Estradiol promotes angiogenesis of bone marrow mesenchymal stem cells by upregulating the PI3K-Akt signaling pathway. Comput Struct Biotechnol J 2022; 20: 3864-3873
  • 26 Latifi-Navid H, Soheili ZS, Samiei S, Sadeghi M, Taghizadeh S, Pirmardan ER, Ahmadieh H. Network analysis and the impact of Aflibercept on specific mediators of angiogenesis in HUVEC cells. J Cell Mol Med 2021; 25: 8285-8299
  • 27 Lam HW, Lin HC, Lao SC, Gao JL, Hong SJ, Leong CW, Yue PYK, Kwan YW, Leung AYH, Wang YT, Lee SMY. The angiogenic effects of Angelica sinensis extract on HUVEC in vitro and zebrafish in vivo. J Cell Biochem 2008; 103: 195-211
  • 28 Jiang M, Murias JM, Chrones T, Sims SM, Lui E, Noble EG. American ginseng acutely regulates contractile function of rat heart. Front Pharmacol 2014; 5: 43
  • 29 Kim JH. Pharmacological and medical applications of Panax ginseng and ginsenosides: a review for use in cardiovascular diseases. J Ginseng Res 2018; 42: 264-269
  • 30 Yao QY, Huang YQ, Liu AD, Zhu MZ, Liu J, Yan H, Zhang QY, Geng B, Gao YS, Du SX, Huang P, Tang CS, Du JB, Jin HF. The vasodilatory effect of sulfur dioxide via SGC/cGMP/PKG pathway in association with sulfhydryl-dependent dimerization. Am J Physiol Regul Integr Comp Physiol 2016; 310: R1073-R1080
  • 31 Zhang K, Song W, Li DL, Jin X. Apigenin in the regulation of cholesterol metabolism and protection of blood vessels. Exp Ther Med 2017; 13: 1719-1724
  • 32 Liu J, Wang XD, Lin JH, Li SH, Deng GX, Wei JR. Classifiers for predicting coronary artery disease based on gene expression profiles in peripheral blood mononuclear cells. Int J Gen Med 2021; 14: 5651-5663
  • 33 Yoodee S, Peerapen P, Plumworasawat S, Thongboonkerd V. ARID1A knockdown in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity. Int J Biol Macromol 2021; 180: 1-13
  • 34 Pulkkinen HH, Kiema M, Lappalainen JP, Toropainen A, Beter M, Tirronen A, Holappa L, Niskanen H, Kaikkonen MU, Ylä-Herttuala S, Laakkonen JP. BMP6/TAZ-Hippo signaling modulates angiogenesis and endothelial cell response to VEGF. Angiogenesis 2021; 24: 129-144
  • 35 Chen SH, Murphy D, Lassoued W, Thurston G, Feldman MD, Lee WMF. Activated STAT3 is a mediator and biomarker of VEGF endothelial activation. Cancer Biol Ther 2008; 7: 1994-2003
  • 36 Yang X, Qiao D, Meyer K, Friedl A. Signal transducers and activators of transcription mediate fibroblast growth hactor-induced vascular endothelial morphogenesis. Cancer Res 2009; 69: 1668-1677
  • 37 Nakhjavani M, Smith E, Townsend AR, Price TJ, Hardingham JE. Anti-Angiogenic Properties of Ginsenoside Rg3. Molecules 2020; 25: 4905
  • 38 Yue PYK, Wong DYL, Wu PK, Leung PY, Mak NK, Yeung HW, Liu L, Cai ZW, Jiang ZH, Fan TPD, Wong RNS. The angiosuppressive effects of 20(R)-ginsenoside Rg3. Biochem Pharmacol 2006; 72: 437-445
  • 39 Che YH, Xu ZR, Ni LL, Dong XX, Yang ZZ, Yang ZB. Isolation and identification of the components in Cybister chinensis Motschulsky against inflammation and their mechanisms of action based on network pharmacology and molecular docking. J Ethnopharmacol 2022; 285: 114851
  • 40 Shi L, Yang F, Luo F, Liu Y, Zhang F, Zou MJ, Liu QZ. Evodiamine exerts anti-tumor effects against hepatocellular carcinoma through inhibiting β-catenin-mediated angiogenesis. Tumor Biol 2016; 37: 12791-12803

Zoom Image
Fig. 1 LC-MS chromatograms of leaf (a), flower (b) and root (c) extracts from P. quinquefolius. The peaks were identified as ginsenoside Re (1), ginsenoside Rb1 (2), pseudoginsenoside F11 (3), ginsenoside Rb2 (4), ginsenoside Rb3 (5), ginsenoside Rd (6), and ginsenoside F2 (7).
Zoom Image
Fig. 2 Pro-angiogenic effects of RE, LE, and FE in transgenic zebrafish (Tg : vegfr2-GFP). The intersegmental vessels are indicated by the white square; Scale bar, 200 µm.
Zoom Image
Fig. 3 Construction of a candidate-target network for ginsenosides against CAD. The triangle nodes represent compounds; the circular nodes and rectangle nodes represent predicted targets and therapeutic targets, respectively.
Zoom Image
Fig. 4 GO enrichment analysis displayed the targets from PPI network are enriched in various biological processes and molecular function.
Zoom Image
Fig. 5 KEGG analysis revealed the top 20 pathway terms associated with the targets from PPI network.
Zoom Image
Fig. 6 The ginsenosides stimulated the proliferation and induced VEGF, FGF2, and p-STAT3 activation in HUVECs. (A) Proliferation effects of ginsenoside F2, (B) Proliferation effects of pseudoginsenoside F11, (C) Ginsenoside F2-mediated alterations in protein expression, (D) Pseudoginsenoside F11-mediated alterations in protein expression. (The experiments were repeated six and three times for cell viability and western blot, respectively; error bars represent means ± SD, *P < 0.05 and ** P < 0.01 vs. control.)