Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273365

ABSTRACT

Though Ginsenoside F2 (GF2), a protopanaxadiol saponin from Panax ginseng, is known to have an anticancer effect, its underlying mechanism still remains unclear. In our model, the anti-glycolytic mechanism of GF2 was investigated in human cervical cancer cells in association with miR193a-5p and the ß-catenin/c-Myc/Hexokinase 2 (HK2) signaling axis. Here, GF2 exerted significant cytotoxicity and antiproliferation activity, increased sub-G1, and attenuated the expression of pro-Poly (ADPribose) polymerase (pro-PARP) and pro-cysteine aspartyl-specific protease (procaspase3) in HeLa and SiHa cells. Consistently, GF2 attenuated the expression of Wnt, ß-catenin, and c-Myc and their downstream target genes such as HK2, pyruvate kinase isozymes M2 (PKM2), and lactate dehydrogenase A (LDHA), along with a decreased production of glucose and lactate in HeLa and SiHa cells. Moreover, GF2 suppressed ß-catenin and c-Myc stability in the presence and absence of cycloheximide in HeLa cells, respectively. Additionally, the depletion of ß-catenin reduced the expression of c-Myc and HK2 in HeLa cells, while pyruvate treatment reversed the ability of GF2 to inhibit ß-catenin, c-Myc, and PKM2 in GF2-treated HeLa cells. Notably, GF2 upregulated the expression of microRNA139a-5p (miR139a-5p) in HeLa cells. Consistently, the miR139a-5p mimic enhanced the suppression of ß-catenin, c-Myc, and HK2, while the miR193a-5p inhibitor reversed the ability of GF2 to attenuate the expression of ß-catenin, c-Myc, and HK2 in HeLa cells. Overall, these findings suggest that GF2 induces apoptosis via the activation of miR193a-5p and the inhibition of ß-catenin/c-Myc/HK signaling in cervical cancer cells.


Subject(s)
Ginsenosides , Hexokinase , MicroRNAs , Proto-Oncogene Proteins c-myc , Signal Transduction , Uterine Cervical Neoplasms , beta Catenin , Humans , Ginsenosides/pharmacology , beta Catenin/metabolism , beta Catenin/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Female , Signal Transduction/drug effects , Hexokinase/metabolism , Hexokinase/genetics , HeLa Cells , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Warburg Effect, Oncologic/drug effects , Apoptosis/drug effects
2.
Phytomedicine ; 129: 155637, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38669969

ABSTRACT

BACKGROUND: Ginsenoside F2 (GF2) serves as the principal intestinal metabolite resulting from the oral intake of Panax ginseng and Panax quinquefolius, exhibiting antioxidative, hypolipidemic, antitumor, and anti-inflammatory properties. Nevertheless, its effect on myocardial infarction (MI) is still unknown. PURPOSE: The purpose of this study is to investigate the protective effect and the underlying mechanisms of GF2 against isoproterenol (ISO)-induced MI. METHODS: ISO-induced H9c2 cardiomyocytes and MI rat models were utilized as in vitro and in vivo models to evaluate the impact of anti-MI of GF2. The underlying mechanisms were investigated using a variety of methodologies, including electrocardiography, Western blot analysis, histopathological examination, immunofluorescence, immunohistochemistry, and ELISA techniques. RESULTS: In vivo experiments, our results indicated that GF2 significantly ameliorated ISO-induced electrocardiographic (ECG) abnormalities, myocardial fiber necrosis, rupture, fibrosis of myocardial tissues, and suppressed cardiac enzyme activities. Meanwhile, GF2 notably raised the activity of antioxidant enzymes like CAT, GSH, and SOD. Furthermore, it downregulated Keap1 expression level while upregulating NQO1, Nrf2, and HO-1 expression levels. Additionally, GF2 suppressed the expression of the cleaved caspase-3 and pro-apoptotic protein Bax while promoting the expression of anti-apoptotic proteins Bcl-2, p-PI3K, and p-Akt. TUNEL fluorescence results also demonstrated that GF2 effectively inhibited cardiomyocyte apoptosis. Furthermore, consistent with the results of animal experiments, GF2 considerably attenuated ROS generation, changed apoptosis and mitochondrial function, and reduced oxidative stress in ISO-induced H9c2 cardiomyocytes through activating Nrf2/HO-1 and PI3K/Akt signaling pathways. CONCLUSION: Taken together, GF2 ameliorated MI by preventing cardiocyte apoptosis, oxidative stress, and mitochondrial dysfunction via modulating the Nrf2/HO-1 and PI3K/Akt signaling pathways, showing potential as a treatment strategy for treating MI.


Subject(s)
Ginsenosides , Isoproterenol , Myocardial Infarction , Myocytes, Cardiac , NF-E2-Related Factor 2 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Animals , Ginsenosides/pharmacology , Myocardial Infarction/chemically induced , Myocardial Infarction/prevention & control , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Rats , Myocytes, Cardiac/drug effects , Apoptosis/drug effects , Oxidative Stress/drug effects , Panax/chemistry , Antioxidants/pharmacology , Cell Line , Heme Oxygenase (Decyclizing)/metabolism , Cardiotonic Agents/pharmacology
3.
J Ginseng Res ; 48(1): 89-97, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38223828

ABSTRACT

Background: Ginsenoside F2 (GF2), the protopanaxadiol-type constituent in Panax ginseng, has been reported to attenuate metabolic dysfunction-associated steatotic liver disease (MASLD). However, the mechanism of action is not fully understood. Here, this study investigates the molecular mechanism by which GF2 regulates MASLD progression through liver X receptor (LXR). Methods: To demonstrate the effect of GF2 on LXR activity, computational modeling of protein-ligand binding, Time-resolved fluorescence resonance energy transfer (TR-FRET) assay for LXR cofactor recruitment, and luciferase reporter assay were performed. LXR agonist T0901317 was used for LXR activation in hepatocytes and macrophages. MASLD was induced by high-fat diet (HFD) feeding with or without GF2 administration in WT and LXRα-/- mice. Results: Computational modeling showed that GF2 had a high affinity with LXRα. LXRE-luciferase reporter assay with amino acid substitution at the predicted ligand binding site revealed that the S264 residue of LXRα was the crucial interaction site of GF2. TR-FRET assay demonstrated that GF2 suppressed LXRα activity by favoring the binding of corepressors to LXRα while inhibiting the accessibility of coactivators. In vitro, GF2 treatments reduced T0901317-induced fat accumulation and pro-inflammatory cytokine expression in hepatocytes and macrophages, respectively. Consistently, GF2 administration ameliorated hepatic steatohepatitis and improved glucose or insulin tolerance in WT but not in LXRα-/- mice. Conclusion: GF2 alters the binding affinities of LXRα coregulators, thereby interrupting hepatic steatosis and inflammation in macrophages. Therefore, we propose that GF2 might be a potential therapeutic agent for the intervention in patients with MASLD.

4.
J Agric Food Chem ; 72(1): 339-350, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38150707

ABSTRACT

Atopic dermatitis (AD) is a complex inflammatory skin disease induced by multiple factors. AD can also cause intestinal inflammation and disorders of the gut microbiota. Ginseng is a kind of edible and medicinal plant; its main active components are ginsenosides. Ginsenosides have a variety of anti-inflammatory effects and regulate the gut microbiota; however, their role in AD and the underlying mechanisms are unclear. In this study, we found that intragastric administration of ginsenoside F2 improved AD-like skin symptoms and reduced inflammatory cell infiltration, serum immunoglobulin E levels, and mRNA expression of inflammatory cytokines in AD mice. 16s rRNA sequencing analysis showed that ginsenoside F2 altered the intestinal microbiota structure and enriched the short-chain fatty acid-producing microbiota in AD mice. Metabolomic analysis revealed that ginsenoside F2 significantly increased the propionic acid (Pa) content of feces and serum in AD mice, which was positively correlated with significant enrichment of Parabacteroides goldsteinii and Lactobacillus plantarum in the intestines. Pa inhibits inflammatory responses in the gut and skin of AD mice through the G-protein-coupled receptor43/NF-κB pathway, thereby improving skin AD symptoms. These results revealed, for the first time, the mechanism by which ginsenoside F2 improves AD through the Pa (a metabolite of intestinal microbiota)-gut-skin axis.


Subject(s)
Dermatitis, Atopic , Gastrointestinal Microbiome , Ginsenosides , Mice , Animals , Dermatitis, Atopic/drug therapy , Ginsenosides/pharmacology , RNA, Ribosomal, 16S
5.
Phytomedicine ; 120: 155031, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37666060

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a common type of cancer that shows great morbidity and mortality rates. However, there are limited available drugs to treat HCC. AIM: The present work focused on discovering the potential anti-HCC compounds from traditional Chinese medicine (TCM) by employing high-throughput sequencing-based high-throughput screening (HTS2) together with the liver cancer pathway-associated gene signature. METHODS: HTS2 assay was adopted for identifying herbs. Protein-protein interaction (PPI) network analysis and computer-aided drug design (CADD) were used to identify key targets and screen the candidate natural products of herbs. Molecular docking, network pharmacology analysis, western blotting, immunofluorescent staining, subcellular fractionation experiment, dual-luciferase reporter gene assay, surface plasmon resonance (SPR) as well as nuclear magnetic resonance (NMR) were performed to validate the ability of compound binding with key target and inhibiting its function. Moreover, cell viability, colony-forming, cell cycle assay and animal experiments were performed to examine the inhibitory effect of compound on HCC. RESULTS: We examined the perturbation of 578 herb extracts on the expression of 84 genes from the liver cancer pathway, and identified the top 20 herbs significantly reverting the gene expression of this pathway. Signal transducer and activator of transcription 3  (STAT3)  was identified as one of the key targets of the liver cancer pathway by PPI network analysis. Then, by analyzing compounds from top 20 herbs utilizing CADD, we found ginsenoside F2 (GF2) binds to STAT3 with high affinity, which was further validated by the results from molecular docking, SPR and NMR. Additionally, our results showed that GF2 suppresses the phosphorylation of Y705 of STAT3, inhibits its nuclear translocation, decreases its transcriptional activity and inhibits the growth of HCC in vitro and in vivo. CONCLUSION: Based on this large-scale transcriptional study, a number of anti-HCC herbs were identified. GF2, a compound derived from TCM, was found to be a chemical basis of these herbs in treating HCC. The present work also discovered that GF2 is a new STAT3 inhibitor, which is able to suppress HCC. As such, GF2 represents a new potential anti-HCC therapeutic strategy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , STAT3 Transcription Factor , Molecular Docking Simulation
6.
Nat Prod Res ; : 1-7, 2023 May 08.
Article in English | MEDLINE | ID: mdl-37157839

ABSTRACT

Ginsenoside F2 is a protopanaxadiol saponin compound with various biological activities, including antioxidant, anti-inflammatory, and anticancer properties. Ginsenoside F2 can be found in ginseng, but in low quantities. Therefore, ginsenoside F2 production predominantly relies on the biotransformation of various ginsenosides, such as ginsenosides Rb1 and Rd. In this study, we reported the production of ginsenoside F2 by gypenoside biotransformation with Aspergillus niger JGL8, isolated from Gynostemma pentaphyllum. Ginsenoside F2 could be produced by two different biotransformation pathways, namely Gyp-V-Rd-F2 and Gyp-XVII-F2. The product exhibited antioxidant activity against free radicals (DPPH) with IC50 value of 29.54 µg/mL. Optimal biotransformation conditions were a pH of 5.0, temperature of 40 °C, and 2 mg/mL of substrate. Enzyme kinetic parameters revealed that the hydrolysis rate of Gyp-V, Rd, and Gyp-XVII was 0.625, 0.588, and 0.417 mM/h, respectively. In conclusion, we demonstrated that gypenoside is a substitutable substrate for ginsenoside F2 biotransformation.

7.
J Ginseng Res ; 47(3): 420-428, 2023 May.
Article in English | MEDLINE | ID: mdl-37252284

ABSTRACT

Background: Ginsenoside F2 (GF2), a minor component of Panax ginseng, has been reported to possess a wide variety of pharmacological activities. However, its effects on glucose metabolism have not yet been reported. Here, we investigated the underlying signaling pathways involved in its effects on hepatic glucose. Methods: HepG2 cells were used to establish insulin-resistant (IR) model and treated with GF2. Cell viability and glucose uptake-related genes were also examined by real-time PCR and immunoblots. Results: Cell viability assays showed that GF2 up to 50 µM did not affect normal and IR-HepG2 cell viability. GF2 reduced oxidative stress by inhibiting phosphorylation of the mitogen-activated protein kinases (MAPK) signaling components such as c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1/2 (ERK1/2), and p38 MAPK, and reducing the nuclear translocation of NF-κB. Furthermore, GF2 activated PI3K/AKT signaling, upregulated the levels of glucose transporter 2 (GLUT-2) and GLUT-4 in IR-HepG2 cells, and promoted glucose absorption. At the same time, GF2 reduced phosphoenolpyruvate carboxykinase and glucose-6-phosphatase expression as well as inhibiting gluconeogenesis. Conclusion: Overall, GF2 improved glucose metabolism disorders by reducing cellular oxidative stress in IR-HepG2 cells via MAPK signaling, participating in the PI3K/AKT/GSK-3ß signaling pathway, promoting glycogen synthesis, and inhibiting gluconeogenesis.

8.
J Ethnopharmacol ; 287: 114927, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-34954265

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Panax ginseng root has been used as tonic in traditional Chinese medicine (TCM) and traditional Japanese Kampo medicine. Steam processing of Panax ginseng root is carried out to enhance its nourishing effects on qi. AIM OF THE STUDY: In order to explore the mechanism of these beneficial effects behind the steam processing of the P. ginseng root, we evaluated effectiveness of processing on the granulocyte-colony stimulating factor (G-CSF) secretion in intestinal epithelial cell-like MCE301 cells. MATERIALS AND METHODS: We collected P. ginseng root samples in the markets of China and Japan. Fresh or dried samples were steamed for different time lengths and subsequently dried and extracted. MCE301 cells were incubated with the medium containing various P. ginseng root extracts, while the concentration of G-CSF in the medium was measured. We also investigated the active ingredients by size exclusion HPLC. RESULTS: The extracts of fresh P. ginseng hairy root samples steamed for more than 6 h significantly induced G-CSF secretion, and the maximum activity was recorded at a 9-h steaming. The same activity was noted when already dried P. ginseng hairy root samples were steamed. The extracts of fresh P. ginseng hairy root without steam processing and those of fresh P. ginseng root body samples with steam processing exhibited no activities. The active ingredients of steamed P. ginseng hairy root samples were high-molecular-weight compounds with an average molecular weight of 758 kDa, and the activity was mediated by the toll-like receptor (TLR) 9. CONCLUSIONS: Our results shed on more light on the mechanism underlying the appearance of immunostimulatory activity of the P. ginseng hairy root induced by steam processing.


Subject(s)
Intestinal Mucosa/drug effects , Panax/chemistry , Plant Extracts/pharmacology , Steam , Animals , Cell Line , Chromatography, High Pressure Liquid , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Intestinal Mucosa/cytology , Mice , Plant Extracts/chemistry , Plant Roots
9.
J Ethnopharmacol ; 282: 114655, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-34537284

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Inflammatory responses are associated wieh the pathophysiology of depression. Ginsenoside Rb1 (Rb1) exerts antidepressant effect, but the relationship between its activity and inflammation remains unclear. AIM OF THE STUDY: In this study, the antidepressant-like effect and underlying mechanisms of Rb1 were been investigated. MATERIALS AND METHODS: The neuroinflammatory mouse model of lipopolysaccharide (LPS)-induced acute depression-like behavior was employed to detect the action of Rb1. An integrative strategy combining the identification of prototype (Rb1) and its metabolites in vivo with network pharmacology analysis was used to explore therapeutic mechanisms of these ingredients. The putative targets and signalings were experimentally validated. The antidepressant-like effect of F2, the metabolite of Rb1, was firstly evaluated. RESULTS: Rb1 significantly ameliorated LPS-induced depressive-like behavior. Rb1 and its metabolites (Rd, F2, compound K, Rh2, Rg3, PPD) were identified and then a disease-component-target network was established. Experimental validation showed that Rb1 inhibited peripheral and hippocampal inflammation via MAPK/NF-κB signaling. In inflammatory-mediated depression state, Rb1 improved impaired glucocorticoid receptor, suppressed indoleamine 2,3-dioxygenase activity, increased 5-HT level and 5-HT1A receptor expression. Additionally, F2 was firstly discovered to exert antidepressant-like effect, and it existed higher activity than Rb1 against depression. CONCLUSION: The study highlighted the potential of Rb1 and F2 as healthy supplement or agent for inflammation-induced depression.


Subject(s)
Depression/drug therapy , Ginsenosides/therapeutic use , Lipopolysaccharides/toxicity , Network Pharmacology , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/therapeutic use , Depression/chemically induced , Ginsenosides/metabolism , Inflammation , Male , Mice , Mice, Inbred ICR
10.
J Agric Food Chem ; 69(32): 9299-9312, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34342980

ABSTRACT

Ginsenoside F2 (GF2) is a protopanaxdiol saponin from Panax ginseng leaves and possesses many potential pharmacological properties. GF2 may prevent obesity by directly binding to the peroxisome proliferator-activated receptor-γ (PPARγ) and inhibiting adipocyte differentiation. However, the mechanism by which GF2 alleviates obesity is unknown. We therefore explored the anti-adipogenesis and anti-obesity effects of GF2 in vitro and in vivo. GF2 inhibited differentiation and reduced the triglyceride (TG) content of 3T3-L1 preadipocytes in the early stage of adipogenesis. Administration of GF2 (50 and 100 mg/kg) to obese mice for 4 weeks reduced the body weight gain, weight of adipose tissues, adipocyte size, and total cholesterol, TG, and AST levels in serum. RNA sequencing and real-time quantitative PCR indicated that GF2 decreased the expression levels of adipokines, including PPARγ, fatty acid synthase, and adiponectin. KEGG enrichment and western blot analyses demonstrated that GF2 accelerated the phosphorylation of AMPK and ACC in vitro and in vivo. Moreover, GF2 promoted the biosynthesis of mitochondria in 3T3-L1 adipocytes and increased the expression of antioxidant enzymes such as SOD and GSH-Px in the liver of obese mice. Therefore, GF2 suppressed adipogenesis and obesity by regulating the expression of adipokines and activating the AMPK pathway. Hence, the findings suggest that GF2 may have potential therapeutic implications to treat obesity.


Subject(s)
Adipogenesis , Anti-Obesity Agents , 3T3-L1 Cells , AMP-Activated Protein Kinases/genetics , Adipogenesis/genetics , Animals , Anti-Obesity Agents/pharmacology , Diet, High-Fat , Ginsenosides , Mice , Mice, Obese , Obesity/drug therapy , Obesity/genetics , PPAR gamma/genetics
11.
Phytomedicine ; 83: 153483, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33578358

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most aggressive tumor residing within the central nervous system, with extremely poor prognosis. Although the cytotoxic effects of ginsenoside F2 (GF2) on GBM were previously suggested, the precise anti-GBM mechanism of GF2 remains unclear. The aim of this study was to explore the anti-cancer molecular mechanism of GF2 toward human GBM. METHODS: GF2-driven cellular toxicity was confirmed in two different GBM cells, U373 and Hs683. To test mitochondrial impairment driven by GF2, we examined the mitochondrial membrane potential, OCR, and ATP production. An intracellular redox imbalance was identified by measuring the relative ratio of reduced glutathione to oxidized glutathione (GSH/GSSG), glutaredoxin (GLRX) mRNA expression, intracellular NAD+ level, and AMPK phosphorylation status. RESULTS: GF2 increased the percentage of cleaved caspase 3-positive cells and γH2AX signal intensities, confirming that GF2 shows the cytotoxicity against GBM. GO enrichment analysis suggested that the mitochondrial function could be negatively influenced by GF2. GF2 reduced the mitochondrial membrane potential, basal mitochondrial respiratory rate, and ATP production capacity. Our results showed that GF2 downregulated the relative GSH/GSSG, intracellular NAD+ level, and GLRX expression, suggesting that GF2 may alter the intracellular redox balance that led to mitochondrial impairment. CONCLUSION: GF2 reduces mitochondrial membrane potential, inhibits cellular oxygen consumption, activates AMPK signaling, and induces cell death. Our study examined the potential vulnerability of mitochondrial activity in GBM, and this may hold therapeutic promise.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Ginsenosides/pharmacology , Glioblastoma/drug therapy , Mitochondria/drug effects , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Glutaredoxins/genetics , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidation-Reduction
12.
Biosci Biotechnol Biochem ; 84(11): 2327-2336, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32698696

ABSTRACT

According to the previously described anti-photoaging effect of the enzyme-processed Panax ginseng extract and Gastrodia elata extract, we hypothesized that the combination of the two extracts would have superior effect to protect human skin from UVB radiation. Besides, the mixture of active components isolated from herbal extracts, ginsenoside F2, and α-gastrodin was investigated on the photo-protective capability. The expression of aging-related markers including matrix metalloproteinase-1 (MMP-1), interleukin-6 (IL-6), and procollagen type 1 was evaluated using ELISA kits. It was reported that the herbal extract at a Panax ginseng extract to Gastrodia elata extract ratio of 1:10 (w/w) and the compound mixture with equal proportion of ginsenoside F2 and α-gastrodin exhibited significant inhibition of MMP-1 and IL-6 production, and marked upregulation of procollagen type 1 formation. Thus, the combination of either the enzyme-processed herbal extracts or their active components would enhance the properties of prevention and treatment of UVB-induced skin damage.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/drug effects , Gastrodia/chemistry , Interleukin-6/metabolism , Matrix Metalloproteinase 1/metabolism , Panax/chemistry , Plant Extracts/pharmacology , Cell Survival/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Radiation-Protective Agents/pharmacology , Skin/cytology , Ultraviolet Rays/adverse effects
13.
Int J Mol Sci ; 21(12)2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32630596

ABSTRACT

A ginsenoside F2-enhanced mixture (SGL 121) increases the content of ginsenoside F2 by biotransformation. In the present study, we investigated the effect of SGL 121 on nonalcoholic fatty liver disease (NAFLD) in vitro and in vivo. High-fat, high-carbohydrate-diet (HFHC)-fed mice were administered SGL 121 for 12 weeks to assess its effect on improving NAFLD. In HepG2 cells, SGL 121 acted as an antioxidant, a hepatoprotectant, and had an anti-lipogenic effect. In NAFLD mice, SGL 121 significantly improved body fat mass; levels of hepatic triglyceride (TG), hepatic malondialdehyde (MDA), serum total cholesterol (TC), high-density lipoprotein (HDL), and low-density lipoprotein (LDL); and activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). In HepG2 cells, induced by oxidative stress, SGL 121 increased cytoprotection, inhibited reactive oxygen species (ROS) production, and increased antioxidant enzyme activity. SGL 121 activated the Nrf2/HO-1 signaling pathway and improved lipid accumulation induced by free fatty acids (FFA). Sterol regulatory element-binding protein-1 (SREBP-1) and fatty acid synthase (FAS) expression was significantly reduced in NAFLD-induced liver and HepG2 cells treated with SGL 121. Moreover, SGL 121 activated adenosine monophosphate-activated protein kinase (AMPK), which plays an important role in the regulation of lipid metabolism. The effect of SGL 121 on the improvement of NAFLD seems to be related to its antioxidant effects and activation of AMPK. In conclusion, SGL 121 can be potentially used for the treatment of NAFLD.


Subject(s)
Ginsenosides/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Diet, High-Fat , Fatty Acids, Nonesterified/metabolism , Ginsenosides/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-846117

ABSTRACT

Objective: Through network pharmacology, the network relationship between the active component of Sanqi Mixture, the target of hepatic ischemia- reperfusion injury(HIRI), and biological pathway was constructed to explore the key target and mechanism of effect of Sanqi Mixture on HIRI. Method: Through literature research at home and abroad, Traditional Chinese Medicine Systems Pharmacology (TCMSP) platform, Pharm Mapper, Swiss Target Prediction and other servers, oral availability (OB) and drug-likeness (DL) were selected as the limited conditions to collect the relevant targets for Sanqi Mixture for intervention in HIRI. The OMIM database was used to screen and collate HIRI related genes and protein targets. Excel table was used to merge and sort the intersection between disease and targets through Cytoscape3.7.2 software plug-ins Network Analyzer, with topological parameters (degree) ≥ 5 (average degrees of freedom 4.5) for the filter to find the core targets; And the intersection targets were imported to the server STRING, and with Confidence Score of 0.85 or higher for the filter conditions to build the core protein interactions (Hub-PPI) network. The intersection target was introduced into FunRich 3.0 software for biological process and biological pathway analysis, and Cytoscape3.7.2 was used to construct the network of "traditional Chinese medicine-active ingredient-HIRI target-biological pathway". Result: Sanqi mixture could reduce the expression of Aspartate aminotransferase (AST) and glutamate transaminase (ALT) in HIRI mice (P < 0.01). After screening, 45 active components of Sanqi Mixture were obtained, corresponding to 3 273 targets, and the main compounds included ursolic acid, oleanolic acid, brucine, quercetin, ginsenoside F2, paeoniflorin, etc. Among the 196 targets obtained by HIRI, 46 targets were intersected with components, including 11-β-hydroxysteroid dehydrogenase (HSD11B1), adenosine receptor A3 (ADORA3), cyclooxygenase 2 (PTGS2), adenosine receptor A1 (ADORA1), protein kinase C-ε (PKC), etc. With the STRING server setting the qualified condition of Confidence Score ≥ 0.85, the PPI network with high Confidence was obtained and clustered into three categories through cluster processing. Five biological processes including protein metabolism, signal transduction, negative regulation of enzyme activity, inflammatory response and transmembrane receptor protein tyrosine kinase signal pathway were analyzed by FunRich software (P < 0.05). 16 biological pathways including integrin-linked kinase signal, TNF receptor signaling pathway, P38 mitogen-activated protein kinase signaling pathway, and TRAIL signaling pathway (P < 0.01). Conclusion: It is preliminarily discussed that Sanqi Mixture intervenes HIRI through the interaction of multiple components and multiple targets, as well as the regulation of multiple biological pathways and biological processes. However, the key core targets and the specific regulation mechanism still need further experimental verification.

15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-846299

ABSTRACT

Objective: To explore the network regulation mechanism of blood-activating and hemostatic and detumescent and analgesic traditional effects of Panax notoginseng. Methods: Targets of the 12 components of P. notoginseng absorbed in plasma were predicted according to the reverse pharmacophore method. Gene ontology (GO) function enrichment and pathway analysis of the targets were analyzed by Omicsbean online analysis software and String 10 database. Finally, Cytoscape software was used to construct the network pharmacology map. Results: A total of 12 compounds (notoginsenoside R1, ginsenoside Rg1, ginsenoside Re, ginsenoside Rh1, ginsenoside Rg2, ginsenoside Rb1, ginsenoside Rd, ginsenoside F2, ginsenoside Rg3, ginsenoside Rk1, dencichine and quercetin) affected 65 pathways through 65 related targets, which were associated with anti-thrombosis, fibrinolysis, angiogenesis, vasodilation, blood coagulation, anti-inflammation and analgesia. The network of "compound-target-pathway-pharmacological action-efficacy" was also constructed. Conclusion: P. notoginseng interferes with multiple biological processes related to activating blood circulation, hemostasis, detumescence and analgesia by acting on several key proteins such as F2, F10, PLAT, VEGFA, NOS2, IL6, PTGES, OPRD1, etc.

16.
Wei Sheng Yan Jiu ; 48(3): 452-457, 2019 May.
Article in Chinese | MEDLINE | ID: mdl-31133133

ABSTRACT

OBJECTIVE: To investigate the inhibitive effects of ginsenoside F2 on oxidative stress in human embryonic kidney cells(HEK-293). METHODS: Hydrogen peroxide induced oxidative stress of HEK-293 cell was used as the research object. HEK-293 cells were pretreated with different concentrations of ginsenoside F2(1.25, 5, 20 µmol/L). Cell viability was measured by MTS assay. Malondialchehyche(MDA) level and activities of antioxidant enzymes(superoxide dismutase SOD, glutathione peroxidase GSH-Px, catalase CAT) were measured by corresponding assay kits. DCFH-DA fluorescent probe assay was used to measure the level of intracellular reactive oxygen species(ROS). Quantitative real-time PCR and Western blot were used to detect the expression of nuclear factor erythroid 2-related factor 2(Nrf2) and kelch-like ECH associated protein 1(Keap1). RESULTS: After treated with 1.25, 5, 20 µmol/L ginsenoside F2, no cytotoxic or proliferative effects were shown on normal HEK-293 cells. After pretreatment with ginsenoside F2, the cell viability was significantly higher than that of the injury group(P<0.05)and increased in a concentration-dependent manner. The fluorescence intensity of oxidative DCF in injured group was significantly increased compared with control group(P<0.05). The fluorescence intensity of cells which pretreated with different concentrations of ginsenoside F2 was gradually weakened(P<0.05). The ROS content of control group was chosen as the standard, and the relative amount of ROS pretreated by ginsenoside F2 decreased in a concentration-dependent manner. After pretreatment of ginsenoside F2, the MDA levels decreased in a concentration-dependent manner and the activities of SOD and GSH-Px were significantly higher than those of the injured group(P<0.05). The activity of CAT was significantly increased with pretreatment of higher concentrations of ginsenoside F2(P<0.05). Furthermore, ginsenoside F2 significantly enhanced the protein and mRNA expressions of Nrf2 and reduced the expressions of Keap1 in a dose-dependent manner(P<0.05). CONCLUSION: Ginsenoside F2 protect HEK-293 cells against H_2O_2-induced oxidative stress through reducing intracellular ROS and MDA, as well as activating Nrf2/Keap1 signaling pathway and antioxidant enzymes.


Subject(s)
Oxidative Stress , Ginsenosides , HEK293 Cells , Humans , Hydrogen Peroxide , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Reactive Oxygen Species
17.
Eur J Pharm Sci ; 134: 116-137, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30981885

ABSTRACT

Autophagy is an evolutionarily conserved catabolic mechanism, by which eukaryotic cells recycle or degrades internal constituents through membrane-trafficking pathway. Thus, autophagy provides the cells with a sustainable source of biomolecules and energy for the maintenance of homeostasis under stressful conditions such as tumor microenvironment. Recent findings revealed a close relationship between autophagy and malignant transformation. However, due to the complex dual role of autophagy in tumor survival or cell death, efforts to develop efficient treatment strategies targeting the autophagy/cancer relation have largely been unsuccessful. Here we review the two-faced role of autophagy in cancer as a tumor suppressor or as a pro-oncogenic mechanism. In this sense, we also review the shared regulatory pathways that play a role in autophagy and malignant transformation. Finally, anti-cancer therapeutic agents used as either inhibitors or inducers of autophagy have been discussed.


Subject(s)
Autophagy/drug effects , Autophagy/physiology , Neoplasms/metabolism , Animals , Antineoplastic Agents , Genes, Tumor Suppressor , Humans , Molecular Targeted Therapy , Neoplasms/therapy , Oncogenes , Tumor Microenvironment
18.
Fitoterapia ; 121: 223-228, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28734737

ABSTRACT

Recently, the allergenicity of ginsenosides, as main active components in ginseng, has attracted much attention. Ginsenoside Rb1 and Rd. have been reported to induce anaphylactoid reaction. In this study, the allergenicity of a series of 20(S)-protopanaxadiol (PPD) type ginsenosides, including Rb1, Rd., F2, Compound K and 20(S)-PPD, was evaluated in rat basophilic leukemia 2H3 (RBL2H3) cells. As a result, 20(S)-PPD had no effect on the mast cell degranulation, but other components showed anaphylactoid potential to different extent. The allergenicity was stronger and stronger according to the order "Rb1, Rd., F2, Compound K". Then, F2 was further verified in RBL-2H3 cells, mouse peritoneal mast cells (MPMCs), Laboratory of Allergic Disease 2 (LAD2) human mast cells in vitro and mice in vivo. Results showed that F2 could induce a significant increase of histamine release and translocation of phosphatidylserine in RBL-2H3 cells. F2 also increased ß-hexosaminidase release and the intracellular Ca2+ concentration of MPMCs and LAD2 cells. In addition, histamine level in serum of mice was elevated dose-dependently. Our study revealed the potential structure-allergenicity relationship of 20(S)-PPD type ginsenosides and first verified the allergenicity of ginsenoside F2. This study could guide the establishment of quality standards for safe application of ginsenoside-containing preparations.


Subject(s)
Ginsenosides/adverse effects , Hypersensitivity/physiopathology , Mast Cells/drug effects , Animals , Cell Line, Tumor , Histamine/metabolism , Humans , Hypersensitivity/metabolism , Male , Mast Cells/metabolism , Mice , Mice, Inbred ICR , Molecular Structure , Phosphatidylserines/metabolism , Rats , beta-N-Acetylhexosaminidases/metabolism
19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-852726

ABSTRACT

Objective: To evaluate the quality of Maiweishen, a simple and accurate HPLC method for determining the contents of 20 active constituents from Maiweishen was established. Methods: The chromatographic separation was achieved on a C18 column (150 mm × 4.6 mm, 5 μm) using a mobile phase made up of acetonitrile and water at a flow rate of 1.0 mL/min. The detection wavelength and column temperature were set as 203 nm and 35 ℃, respectively. Results: Sixteen ginsenosides (Rg1, Re, Rf, Rb1, Rg2, Rc, Rb2, Rb3, F1, Rd, F2, Rg3, protopanaxatriol, compounds K, Rh2, and protopanaxadiol), three kinds of lignan in Schisandra chinensis (schizandrol A, schizandrin A, B), and ophiopogonin D were separated at baseline with good linearity (r ≥ 0.999 6). The recovery rates were 96%-102% (RSD < 2%). Conclusion: The method is simple, fast, accurate, and could be applied to the quality control of Maiweishen.

20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-852943

ABSTRACT

Objective: To establish an HPLC fingerprint of Qinqi Rheumatism Formula (QRF) for its quality control and effective components determination. Methods: HPLC method was performed on Inert Sustain C18 (150 mm × 4.6 mm, 5 μm) column with gradient elution composed of acetonitrile-aqueous solution containing 0.1% phosphoric acid at the flow rate of 1.0 mL/min. The column temperature was set at 30℃, while the detective wavelength was set at 203 nm. The common mode of HPLC fingerprint for 10 batches of QRF was established with Similarity Evaluation System for Chromatographic Fingerprint of Traditional Chinese Medicine (2004 A edition) and the common peaks were identified by reference compounds. Results: Fingerprints of QRF were established. The similarities of the 10 batches of samples were above 0.99. A total of 19 common peaks were found. Eight mutual peaks were from Panacis Majoris Rhizoma, eight mutual peaks were from Gentianae Macrophyllae Radix, and five mutual peaks were from Corni Fructus (Peak 1 was the common peak from Panacis Majoris Rhizoma, Gentianae Macrophyllae Radix, and Corni Fructus). Based on the retention time of reference substances, eight constituents including loganic acid (peak 2), morroniside (peak 3), gentiopicroside (peak 5), loganin (peak 6), ginsenoside Ro (peak 12), ginsenoside F1 (peak 13), panax japonicus IVa (peak 14), and ginsenoside F2 (peak 17) were indentified. Conclusion: The method is stable, specific, and reproducible, and can be used for the quality control of QRF and the study of its effective components.

SELECTION OF CITATIONS
SEARCH DETAIL