1
|
Bruggink V, Gutjahr C, Decker A, Engelbrecht H, Beekmann U, Kralisch D, Werner M, Schädel P, Jordan PM, Werz O, Hofstetter RK. Ginsenosides from Panax ginseng modulate lipid mediator profiles in human leukocytes by interference with cellular 5-lipoxygenase activity. Biochem Pharmacol 2025; 236:116882. [PMID: 40118289 DOI: 10.1016/j.bcp.2025.116882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/07/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Lipid mediators are a superfamily of bioactive molecules that are crucially involved in immune responses, regulating all stages of inflammation. Panax (P.) ginseng has pleiotropic pharmacological effects, including anti-cancer, anti-diabetic, and anti-inflammatory properties. Ginsenosides, unique triterpenoid glycosides from the plant's root, are proposed as active ingredients responsible for the immunomodulating potential of P.ginseng. Here, we comprehensively screened 23 ginsenosides for manipulating the lipid mediator network in various primary human innate immune cells. Several ginsenosides selectively inhibited 5-lipoxygenase (5-LOX)-mediated formation of pro-inflammatory leukotriene B4, but not of prostaglandins, in monocyte-derived macrophages and polymorphonuclear leukocytes by a unique irreversible mechanism. Structure-activity relationships revealed (i) higher anti-5-LOX activity of PPD-type ginsenosides, (ii) correlation with lipophilicity (R2 = 0.91), and (iii) eudysmic ratios favoring the 20S-epimers. Our findings highlight ginsenosides as immunomodulatory principles of P. ginseng and reveal abrogation of leukotriene formation rather than interference with prostaglandins as immediate anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Vera Bruggink
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany; JeNaCell GmbH-An Evonik Company, 07745 Jena, Germany
| | - Clemens Gutjahr
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Angelika Decker
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Hannes Engelbrecht
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Uwe Beekmann
- JeNaCell GmbH-An Evonik Company, 07745 Jena, Germany
| | - Dana Kralisch
- JeNaCell GmbH-An Evonik Company, 07745 Jena, Germany; Evonik Operations GmbH, 45128 Essen, Germany
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Patrick Schädel
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany
| | - Robert K Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743 Jena, Germany.
| |
Collapse
|
2
|
Wang Y, Han Q, Zhang S, Xing X, Sun X. New perspective on the immunomodulatory activity of ginsenosides: Focus on effective therapies for post-COVID-19. Biomed Pharmacother 2023; 165:115154. [PMID: 37454595 DOI: 10.1016/j.biopha.2023.115154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
More than 700 million confirmed cases of Coronavirus Disease-2019 (COVID-19) have been reported globally, and 10-60% of patients are expected to exhibit "post-COVID-19 symptoms," which will continue to affect human life and health. In the absence of safer, more specific drugs, current multiple immunotherapies have failed to achieve satisfactory efficacy. Ginseng, a traditional Chinese medicine, is often used as an immunomodulator and has been used in COVID-19 treatment as a tonic to increase blood oxygen saturation. Ginsenosides are the main active components of ginseng. In this review, we summarize the multiple ways in which ginsenosides affect post-COVID-19 symptoms, including inhibition of lipopolysaccharide, tumor necrosis factor signaling, modulation of chemokine receptors and inflammasome activation, induction of macrophage polarization, effects on Toll-like receptors, nuclear factor kappa-B, the mitogen-activated protein kinase pathway, lymphocytes, intestinal flora, and epigenetic regulation. Ginsenosides affect virus-mediated tissue damage, local or systemic inflammation, immune modulation, and other links, thus alleviating respiratory and pulmonary symptoms, reducing the cardiac burden, protecting the nervous system, and providing new ideas for the rehabilitation of patients with post-COVID-19 symptoms. Furthermore, we analyzed its role in strengthening body resistance to eliminate pathogenic factors from the perspective of ginseng-epidemic disease and highlighted the challenges in clinical applications. However, the benefit of ginsenosides in modulating organismal imbalance post-COVID-19 needs to be further evaluated to better validate the pharmacological mechanisms associated with their traditional efficacy and to determine their role in individualized therapy.
Collapse
Affiliation(s)
- Yixin Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Qin Han
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Shuxia Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| |
Collapse
|
3
|
Lin H, Wang C, Yu H, Liu Y, Tan L, He S, Li Z, Wang C, Wang F, Li P, Liu J. Protective effect of total Saponins from American ginseng against cigarette smoke-induced COPD in mice based on integrated metabolomics and network pharmacology. Biomed Pharmacother 2022; 149:112823. [PMID: 35334426 DOI: 10.1016/j.biopha.2022.112823] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/02/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory disease. Aiming at assessing the effect of total saponins from American ginseng on COPD, both the chemical composition and anti-COPD activity of total saponins from wild-simulated American ginseng (TSW) and field-grown American ginseng (TSF) were investigated in this study. Firstly, a HPLC-ELSD chromatographic method was established to simultaneously determine the contents of 22 saponins in TSW and TSF. Secondly, CS-induced COPD mouse model was established to evaluate the activity of TSW and TSF. The results indicated that both TSW and TSF had the protective effect against COPD by alleviating oxidative stress and inflammatory response. TSW showed a stronger effect than TSF. Thirdly, an integrated approach involving metabolomics and network pharmacology was used to construct the "biomarker-reaction-enzyme-target" correlation network aiming at further exploring the observed effects. As the results, 15 biomarkers, 9 targets and 5 pathways were identified to play vital roles in the treatment of TSW and TSF on COPD. Fourthly, based on network pharmacology and the CS-stimulated A549 cell model, ginsenoside Rgl, Rc, oleanolic acid, notoginsenoside R1, Fe, silphioside B were certified to be the material basis for the stronger effect of TSW than TSF. Finally, the molecular docking were performed to visualize the binding modes. Our findings suggested that both TSW and TSF could effectively ameliorate the progression of COPD and might be used for the treatment of COPD.
Collapse
Affiliation(s)
- Hongqiang Lin
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Caixia Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hui Yu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yunhe Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Luying Tan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shanmei He
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuoqiao Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China
| | - Fang Wang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China.
| |
Collapse
|
4
|
Huynh DTN, Jin Y, Myung CS, Heo KS. Ginsenoside Rh1 Induces MCF-7 Cell Apoptosis and Autophagic Cell Death through ROS-Mediated Akt Signaling. Cancers (Basel) 2021; 13:cancers13081892. [PMID: 33920802 PMCID: PMC8071122 DOI: 10.3390/cancers13081892] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Breast cancer (BC) is the most common cause of cancer-related deaths among women worldwide, and its incidence has been increasing. However, current therapeutic approaches, such as chemotherapy, radiation, and hormonal therapy, have become increasingly ineffective because of their severe adverse effects and multidrug resistance. Therefore, the discovery of new potential candidates for BC therapy is essential. Here, we investigated whether ginsenoside Rh1 exhibits anticancer effects on BC. We found that this ginsenoside effectively inhibited the growth of BC cells in both cell cultures and mice. Therefore, ginsenoside Rh1 is a promising candidate for BC treatment. Abstract Breast cancer (BC) is the leading cause of cancer-related deaths among women worldwide. Ginsenosides exhibit anticancer activity against various cancer cells. However, the effects of ginsenoside Rh1 on BC and the underlying mechanisms remain unknown. Here, we investigated the anticancer effects of Rh1 on human BC MCF-7 and HCC1428 cells and the underlying signaling pathways. The anticancer effects of Rh1 in vitro were evaluated using sulforhodamine B (SRB), 3-(4, 5-dimethylthiazole-2-yl)-2, 5-diphenyltetrazolium bromide (MTT), clonogenic assay, propidium iodide (PI)/Hoechst staining, Western blotting, flow cytometry, and immunofluorescence analysis. The in vivo effects of Rh1 were determined using a xenograft model via hematoxylin and eosin and the immunohistochemistry staining of tumor tissues. We found that Rh1 exerted cytotoxicity in the cells by increasing cell apoptosis, autophagy, and cell cycle arrest. These effects were further enhanced by a phosphatidylinositol 3-kinase (PI3K) inhibitor but were rescued by the inhibition of reactive oxygen species (ROS). Moreover, enhanced ROS generation by Rh1 inhibited the activation of the PI3K/Akt pathway. Consistently, Rh1 treatment significantly reduced tumor growth in vivo and increased the ROS production and protein expression of LC3B and cleaved caspase-3 but decreased the phosphorylation of Akt and retinoblastoma (Rb) in tumor tissues. Taken together, Rh1 exerted a potential anticancer effect on BC cells by inducing cell cycle arrest, apoptosis, and autophagy via inhibition of the ROS-mediated PI3K/Akt pathway.
Collapse
Affiliation(s)
- Diem Thi Ngoc Huynh
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (C.-S.M.)
- Department of Pharmacy, Da Nang University of Medical Technology and Pharmacy, Da Nang 550000, Vietnam
| | - Yujin Jin
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (C.-S.M.)
| | - Chang-Seon Myung
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (C.-S.M.)
| | - Kyung-Sun Heo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (C.-S.M.)
- Correspondence: ; Tel.: +82-42-821-5927
| |
Collapse
|
5
|
Ginsenoside Rh1 Alleviates HK-2 Apoptosis by Inhibiting ROS and the JNK/p53 Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3401067. [PMID: 32695207 PMCID: PMC7362279 DOI: 10.1155/2020/3401067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022]
Abstract
Background Cisplatin is widely used in the treatment of malignant patients; however, its adverse nephrotoxic effects limit its clinical use. Ginsenoside Rh1 is a main component of ginseng and has many pharmaceutical effects, including immunomodulatory effects. Objective The objective of this research is to assess the effects of ginsenoside Rh1 on a cisplatin-induced HK-2 injury model and to study its potential effect mechanisms. Methods HK-2 cell vitality was assessed via Cell Counting Kit-8 (CCK-8) assay. Carboxyfluorescein succinimidyl ester/propidium iodide (CFSF/PI) staining was used to detect the apoptosis of HK-2 cells. ROS expression was detected by DCFDA. The expressions of JNK, p53, caspase-3, Bax, and NGAL were detected by western blot. Results Ginsenoside Rh1 was found to increase the vitality of HK-2 cells and inhibit ROS production and the apoptosis of HK-2 cells in a cisplatin-induced injury model. Ginsenoside Rh1 was found to inhibit the expression of JNK, p53, caspase-3, Bax, and NGAL in a cisplatin-induced injury model. Conclusion Ginsenoside Rh1 alleviated HK-2 apoptosis in a cisplatin-induced injury model by inhibiting ROS production and the JNK/p53 pathway. Ginsenoside Rh1 may be a promising drug for the alleviation of cisplatin-induced nephrotoxicity in malignant patients.
Collapse
|
6
|
Ge S, Liu W, Liu H, Liu F, Yu J, Yan M, Huang J. Colorimetric detection of the flux of hydrogen peroxide released from living cells based on the high peroxidase-like catalytic performance of porous PtPd nanorods. Biosens Bioelectron 2015; 71:456-462. [DOI: 10.1016/j.bios.2015.04.055] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 11/25/2022]
|
7
|
Shergis JL, Di YM, Zhang AL, Vlahos R, Helliwell R, Ye JM, Xue CC. Therapeutic potential of Panax ginseng and ginsenosides in the treatment of chronic obstructive pulmonary disease. Complement Ther Med 2014; 22:944-53. [PMID: 25440386 DOI: 10.1016/j.ctim.2014.08.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/06/2014] [Accepted: 08/10/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a major global health burden and will become the third largest cause of death in the world by 2030. It is currently believed that an exaggerated inflammatory response to inhaled irritants, in particular cigarette smoke, cause progressive airflow limitation. This inflammation, where macrophages, neutrophils and lymphocytes are prominent, leads to oxidative stress, emphysema, airways fibrosis and mucus hypersecretion. COPD responds poorly to current anti-inflammatory treatments including corticosteroids, which produce little or no benefit. Panax ginseng has a long history of use in Chinese medicine for respiratory conditions, including asthma and COPD. OBJECTIVES In this perspective we consider the therapeutic potential of Panax ginseng for the treatment of COPD. RESULTS Panax ginseng and its compounds, ginsenosides, have reported effects through multiple mechanisms but primarily have anti-inflammatory and anti-oxidative effects. Ginsenosides are functional ligands of glucocorticoid receptors and appear to inhibit kinase phosphorylation including MAPK and ERK1/2, NF-κB transcription factor induction/translocation, and DNA binding. They also inhibit pro-inflammatory mediators, TNF-α, IL-6, IL-8, ROS, and proteases such as MMP-9. Panax ginseng protects against oxidative stress by increasing anti-oxidative enzymes and reducing the production of oxidants. CONCLUSION Given that Panax ginseng and ginsenosides appear to inhibit processes related to COPD pathogenesis, they represent an attractive therapeutic target for the treatment of COPD.
Collapse
Affiliation(s)
- J L Shergis
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia
| | - Y M Di
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia
| | - A L Zhang
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia
| | - R Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia
| | - R Helliwell
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia
| | - J M Ye
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia
| | - C C Xue
- Traditional and Complementary Medicine Program, School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
8
|
Mi J, Zhang M, Zhang H, Wang Y, Wu S, Hu P. Coupling of ultrasound-assisted extraction and expanded bed adsorption for simplified medicinal plant processing and its theoretical model: extraction and enrichment of ginsenosides from Radix Ginseng as a case study. J Sep Sci 2013; 36:593-601. [PMID: 23341270 DOI: 10.1002/jssc.201200745] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 09/22/2012] [Accepted: 10/05/2012] [Indexed: 11/07/2022]
Abstract
A high-efficient and environmental-friendly method for the preparation of ginsenosides from Radix Ginseng using the method of coupling of ultrasound-assisted extraction with expanded bed adsorption is described. Based on the optimal extraction conditions screened by surface response methodology, ginsenosides were extracted and adsorbed, then eluted by the two-step elution protocol. The comparison results between the coupling of ultrasound-assisted extraction with expanded bed adsorption method and conventional method showed that the former was better than the latter in both process efficiency and greenness. The process efficiency and energy efficiency of the coupling of ultrasound-assisted extraction with expanded bed adsorption method rapidly increased by 1.4-fold and 18.5-fold of the conventional method, while the environmental cost and CO(2) emission of the conventional method were 12.9-fold and 17.0-fold of the new method. Furthermore, the theoretical model for the extraction of targets was derived. The results revealed that the theoretical model suitably described the process of preparing ginsenosides by the coupling of ultrasound-assisted extraction with expanded bed adsorption system.
Collapse
Affiliation(s)
- Jianing Mi
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, P R China
| | | | | | | | | | | |
Collapse
|
9
|
Ryu JS, Lee HJ, Bae SH, Kim SY, Park Y, Suh HJ, Jeong YH. The bioavailability of red ginseng extract fermented by Phellinus linteus. J Ginseng Res 2013; 37:108-16. [PMID: 23717164 PMCID: PMC3659629 DOI: 10.5142/jgr.2013.37.108] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 09/25/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
For the improvement of ginsenoside bioavailability, the ginsenosides of fermented red ginseng by Phellinus linteus (FRG) were examined with respect to bioavailability and physiological activity. The polyphenol content of FRG (19.14±0.50 mg/g) was significantly higher (p<0.05) compared with that of non-fermented red ginseng (NFRG, 11.31±1.15 mg/g). The antioxidant activities in FRG, such as 2,2’-diphenyl-1-picrylhydrazyl, 2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid, and ferric reducing antioxidant power, were significantly higher (p<0.05) than those in NFRG. The HPLC analysis results showed that the FRG had a high level of ginsenoside metabolites. The total ginsenoside contents in NFRG and FRG were 41.65±1.53 mg/g and 50.12±1.43 mg/g, respectively. However, FRG had a significantly higher content (33.90±0.97 mg/g) of ginsenoside metabolites (Rg3, Rg5, Rk1, compound K, Rh1, F2, and Rg2) compared with NFRG (14.75±0.46 mg/g). The skin permeability of FRG was higher than that of NFRG using Franz diffusion cell models. In particular, after 3 h, the skin permeability of FRG was significantly higher (p<0.05) than that of NFRG. Using a rat everted intestinal sac model, FRG showed a high transport level compared with NFRG after 1 h. FRG had dramatically improved bioavailability compared with NFRG as indicated by skin permeation and intestinal permeability. The significantly greater bioavailability of FRG may have been due to the transformation of its ginsenosides by fermentation to more easily absorbable forms (ginsenoside metabolites).
Collapse
Affiliation(s)
- Jae Sik Ryu
- Department of Food Science and Nutrition, Dankook University, Yongin 448-701, Korea
| | | | | | | | | | | | | |
Collapse
|
10
|
Qian ZJ, Jung WK, Kang KH, Ryu B, Kim SK, Je JY, Heo SJ, Oh C, Kang DH, Park WS, Choi IW. IN VITRO ANTIOXIDANT ACTIVITIES OF THE FERMENTED MARINE MICROALGA PAVLOVA LUTHERI (HAPTOPHYTA) WITH THE YEAST HANSENULA POLYMORPHA(1). JOURNAL OF PHYCOLOGY 2012; 48:475-482. [PMID: 27009737 DOI: 10.1111/j.1529-8817.2012.01117.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Microalgae are major primary producers of organic matter in aquatic environments through their photosynthetic activities. Fermented microalga (Pavlova lutheri Butcher) preparation (FMP) is the product of yeast fermentation by Hansenula polymorpha. It was tested for the antioxidant activities including lipid peroxidation inhibitory activity, free-radical-scavenging activity, inhibition of reactive oxygen species (ROS) on mouse macrophages (RAW264.7 cell), and inhibited myeloperoxidase (MPO) activity in human myeloid cells (HL60). FMP exhibited the highest antioxidant activity on free-radical scavenging, inhibitory intracellular ROS, and inhibited MPO activity. MTT [3-(4,5-dimethyl-2-yl)-2,5-diphenyltetrazolium bromide] assay showed no cytotoxicity in mouse macrophages (RAW264.7 cell), human myeloid cells (HL60), and human fetal lung fibroblast cell line (MRC-5). Furthermore, the antioxidative mechanism of FMP was evaluated by protein expression levels of antioxidant enzyme (superoxide dismutase [SOD] and glutathione [GSH]) using Western blot. The results obtained in the present study indicated that FMP is a potential source of natural antioxidant.
Collapse
Affiliation(s)
- Zhong-Ji Qian
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Won-Kyo Jung
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Kyong-Hwa Kang
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - BoMi Ryu
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Se-Kwon Kim
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Jae-Young Je
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Soo-Jin Heo
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Chulhong Oh
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Do-Hyung Kang
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Won Sun Park
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Il-Whan Choi
- Department of Marine Life Science and Marine Life Research and Education Center, Chosun University, Gwangju 501-759 and Wando 537-863, KoreaDepartment of Chemistry, Pukyong National University, Busan 608-737, KoreaSchool of Food Technology and Nutrition, Chonnam National University, Yeosu 550-749, KoreaKorea Ocean Research and Development Institute (KORDI), Ansan, P.O. BOX 29, Seoul 426-744, KoreaDepartment of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South KoreaDepartment of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| |
Collapse
|
11
|
Effects of five oleanolic acid triterpenoid saponins from the rhizome of Anemone raddeana on stimulus-induced superoxide generation, phosphorylation of proteins and translocation of cytosolic compounds to cell membrane in human neutrophils. Fitoterapia 2012; 83:402-7. [DOI: 10.1016/j.fitote.2011.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/30/2011] [Accepted: 12/01/2011] [Indexed: 11/21/2022]
|
12
|
Wu P, Qian Y, Du P, Zhang H, Cai C. Facile synthesis of nitrogen-doped graphene for measuring the releasing process of hydrogen peroxide from living cells. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm16929k] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
13
|
Bae SH, Lee HS, Kim MR, Kim SY, Kim JM, Suh HJ. Changes of ginsenoside content by mushroom mycelial fermentation in red ginseng extract. J Ginseng Res 2011; 35:235-42. [PMID: 23717066 PMCID: PMC3659518 DOI: 10.5142/jgr.2011.35.2.235] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 02/28/2011] [Accepted: 03/16/2011] [Indexed: 11/28/2022] Open
Abstract
To obtain microorganisms for the microbial conversion of ginsenosides in red ginseng extract (RGE), mushroom mycelia were used for the fermentation of RGE. After fermentation, total sugar contents and polyohenol contents of the RGEs fermented with various mushrooms were not a significant increase between RGE and the ferments. But uronic acid content was relatively higher in the fermented RGEs cultured with Lentus edodes (2155.6 μg/mL), Phelllinus linteus (1690.9 μg/mL) and Inonotus obliquus 26137 and 26147 (1549.5 and 1670.7 μg/mL) compared to the RGE (1307.1 μg/mL). The RGEs fermented by Ph. linteus, Cordyceps militaris, and Grifola frondosa showed particularly high levels of total ginsenosides (20018.1, 17501.6, and 16267.0 μg/mL, respectively). The ferments with C. militaris (6974.2 μg/mL), Ph. linteus (9109.2 μg/mL), and G. frondosa (7023.0 μg/mL) also showed high levels of metabolites (sum of compound K, Rh1, Rg5, Rk1, Rg3, and Rg2) compared to RGE (3615.9 μg/mL). Among four different RGE concentrations examined, a 20 brix concentration of RGE was favorable for the fermentation of Ph. linteus. Maximum biotransformation of ginsneoside metabolites (9395.5 μg/mL) was obtained after 5 days fermentation with Ph. linteus. Maximum mycelial growth of 2.6 mg/mL was achieved at 9 days, in which growth was not significantly different during 5 to 9 days fermentation. During fermentation of RGE by Ph. linteus in a 7 L fermenter, Rg3, Rg5, and Rk1 contents showed maximum concentrations after 5 days similar to flask fermentation. These results confirm that fermentation with Ph. linteus is very useful for preparing minor ginsenoside metabolites while being safe for foods.
Collapse
Affiliation(s)
- Song Hwan Bae
- Department of Food and Biotechnology, Hankyong National University, Anseong 456-749, Korea
| | | | | | | | | | | |
Collapse
|
14
|
Wu P, Cai Z, Chen J, Zhang H, Cai C. Electrochemical measurement of the flux of hydrogen peroxide releasing from RAW 264.7 macrophage cells based on enzyme-attapulgite clay nanohybrids. Biosens Bioelectron 2011; 26:4012-7. [DOI: 10.1016/j.bios.2011.03.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/18/2011] [Accepted: 03/19/2011] [Indexed: 12/28/2022]
|
15
|
Wei S, Chen G, He W, Chi H, Abe H, Yamashita K, Yokoyama M, Kodama H. Inhibitory effects of secoiridoids from the roots of Gentiana straminea on stimulus-induced superoxide generation, phosphorylation and translocation of cytosolic compounds to plasma membrane in human neutrophils. Phytother Res 2011; 26:168-73. [PMID: 21584870 DOI: 10.1002/ptr.3496] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 11/10/2022]
Abstract
Gentiana straminea Maxim. has been used widely as a traditional Chinese medicine for the treatment of rheumarthritis, icterepatitis, constipation, pain and hypertension. Five secoiridoids, gentiopicroside (GTP), 6'-O-(2-hydroxy-3-O-β-D-glucopyranosyl-benzoyl)-sweroside (HGBS), 6'-O-β-D-glucosylgentiopicroside (GGTP), sweroside (SW) and swertiamarin (STM) were isolated from the roots of G. straminea. The effect of these secoiridoids on stimulus-induced superoxide generation in human neutrophils was assayed by measuring the reduction of ferricytochrome c. Tyrosyl or serine/threonine phosphorylation of neutrophil proteins, and translocation of the cytosolic compounds to the cell membrane were also investigated using specific monoclonal antibodies. The five secoiridoids used in the present experiment suppressed N-formyl-methionyl-leucyl-phenylalanine (fMLP)-induced superoxide generation in a concentration dependent manner. GTP and HGBS also suppressed phorbol 12-myristate 13-acetate (PMA) and arachidonic acid (AA)-induced superoxide generation. However, the other three secoiridoids showed no effect on PMA- and AA-induced superoxide generation. fMLP-, PMA- and AA-induced tyrosyl or serine/threonine phosphorylation and translocation of the cytosolic proteins to the cell membrane were suppressed in parallel with the suppression of the stimulus-induced superoxide generation.
Collapse
Affiliation(s)
- Shihu Wei
- Department of Anesthesiology and Critical Care Medicine, Kochi Medical School, Nankoku-Shi, Kochi 783-8505, Japan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Xu C, Teng J, Chen W, Ge Q, Yang Z, Yu C, Yang Z, Jia W. 20(S)-protopanaxadiol, an active ginseng metabolite, exhibits strong antidepressant-like effects in animal tests. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:1402-11. [PMID: 20647027 DOI: 10.1016/j.pnpbp.2010.07.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 07/10/2010] [Accepted: 07/12/2010] [Indexed: 12/21/2022]
Abstract
Ginseng has been used for mood adjustment in traditional Chinese medicine for thousands of years. Our previous study has shown that, total ginsenosides, the major pharmacologically functional ingredients of ginseng, possess antidepressant activity. In the present study, we hypothesized that an intestinal metabolite of ginseng, 20(S)-protopanaxadiol (code name S111), as a post metabolism compound (PMC) of ingested ginsenosides, may be responsible for the antidepressant activity of ginseng. To test this hypothesis, antidepressant-like activity of orally given S111 was measured in animal tests including tail suspension test, forced swimming test and rat olfactory bulbectomy depression model. In all those tests, S111 demonstrated antidepressant-like activity as potent as fluoxetine. S111 treated bulbectomy animals had higher levels of monoamine neurotransmitters in the brain and in vitro reuptake assay showed that S111 had a mild inhibitory effect. Furthermore, S111 but not fluoxetine significantly reduced brain oxidative stress and down-regulated serum corticosterone concentration in bulbectomy animals. No disturbance to central nervous system (CNS) normal functions were found in S111 treated animals. These results suggest that the ginseng active metabolite S111 is a potential antidepressant. Since the monoamine reuptake activity of this compound is rather weak, it remains to be investigated whether its antidepressant-like effect is by mechanisms that are different from current antidepressants. Furthermore, this study has demonstrated that post metabolism compounds (PMCs) of herb medicines such as S111 may be a novel source for drug discovery from medicinal herbs.
Collapse
Affiliation(s)
- Changjiang Xu
- Shanghai Innovative Research Centre of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Li YX, Li Y, Lee SH, Qian ZJ, Kim SK. Inhibitors of oxidation and matrix metalloproteinases, floridoside, and D-isofloridoside from marine red alga Laurencia undulata. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:578-586. [PMID: 20017487 DOI: 10.1021/jf902811j] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
In the exploration of abundant marine biological resources, edible red alga Laurencia undulata led to two bioactive isolates: floridoside (1) and D-isofloridoside (2). For the first time, the antioxidant properties of both derivatives (1 and 2) were characterized via free radical scavenging using the ESR technique, reactive oxygen species (ROS) inhibition, membrane protein oxidation, myeloperoxidase (MPO) inhibition, gene expression levels of glutathione (GSH) and superoxide dismutase (SOD), and protein expression of MMP-2 and MMP-9. The results demonstrate that floridoside and D-isofloridoside possess significant antioxidant capacity and are potential inhibitors of MMP-2 and MMP-9. These results clarified that these components may be responsible for the relative activities of crude extract from this genus, which is used as folk medicine. Furthermore, the structure-activity relationships were also suggested. Both isomers could be effective candidates for applications in food and pharmaceutical fields as natural marine antioxidants.
Collapse
Affiliation(s)
- Yong-Xin Li
- Department of Chemistry, Pukyong National University, Busan 608-737, Republic of Korea
| | | | | | | | | |
Collapse
|