51
|
Xu J, Chen HB, Li SL. Understanding the Molecular Mechanisms of the Interplay Between Herbal Medicines and Gut Microbiota. Med Res Rev 2017; 37:1140-1185. [PMID: 28052344 DOI: 10.1002/med.21431] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/21/2016] [Accepted: 11/16/2016] [Indexed: 02/06/2023]
Abstract
Herbal medicines (HMs) are much appreciated for their significant contribution to human survival and reproduction by remedial and prophylactic management of diseases. Defining the scientific basis of HMs will substantiate their value and promote their modernization. Ever-increasing evidence suggests that gut microbiota plays a crucial role in HM therapy by complicated interplay with HM components. This interplay includes such activities as: gut microbiota biotransforming HM chemicals into metabolites that harbor different bioavailability and bioactivity/toxicity from their precursors; HM chemicals improving the composition of gut microbiota, consequently ameliorating its dysfunction as well as associated pathological conditions; and gut microbiota mediating the interactions (synergistic and antagonistic) between the multiple chemicals in HMs. More advanced experimental designs are recommended for future study, such as overall chemical characterization of gut microbiota-metabolized HMs, direct microbial analysis of HM-targeted gut microbiota, and precise gut microbiota research model development. The outcomes of such research can further elucidate the interactions between HMs and gut microbiota, thereby opening a new window for defining the scientific basis of HMs and for guiding HM-based drug discovery.
Collapse
Affiliation(s)
- Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, P.R. China.,Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, P.R. China
| |
Collapse
|
52
|
Kim MG, Kim Y, Jeon JY, Kim DS. Effect of fermented red ginseng on cytochrome P450 and P-glycoprotein activity in healthy subjects, as evaluated using the cocktail approach. Br J Clin Pharmacol 2016; 82:1580-1590. [PMID: 27495955 PMCID: PMC5099554 DOI: 10.1111/bcp.13080] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/22/2016] [Accepted: 08/01/2016] [Indexed: 11/28/2022] Open
Abstract
Aims We assessed the drug interaction profile of fermented red ginseng with respect to the activity of major cytochrome (CYP) P450 enzymes and of a drug transporter protein, P‐glycoprotein (P‐gp), in healthy volunteers. Methods This study was an open‐label crossover study. The CYP probe cocktail drugs caffeine, losartan, dextromethorphan, omeprazole, midazolam and fexofenadine were administered before and after 2 weeks of fermented red ginseng administration. Plasma samples were collected, and tolerability was assessed. Pharmacokinetic parameters were calculated, and the 90% confidence intervals (CIs) of the geometric mean ratios of the parameters were determined from logarithmically transformed data. Values were compared between before and after fermented red ginseng administration using analysis of variance (anova). Results Fifteen healthy male subjects were evaluated, none of whom were genetically defined as a poor CYP2C9, CYP2C19 or CYP2D6 metabolizer based on genotyping. Before and after fermented red ginseng administration, the geometric least‐square mean metabolic ratio (90% CI) was 0.901 (0.830–0.979) for caffeine (CYP1A2) to paraxanthine, 0.774 (0.720–0.831) for losartan (CYP2C9) to EXP3174, 1.052 (0.925–1.197) for omeprazole (CYP2C19) to 5‐hydroxyomeprazole, 1.150 (0.860–1.538) for dextromethorphan (CYP2D6) to dextrorphan, and 0.816 (0.673–0.990) for midazolam (CYP3A4) to 1‐hydroxymidazolam. The geometric mean ratio of the area under the curve of the last sampling time (AUClast) for fexofenadine (P‐gp) was 1.322 (1.112–1.571). Conclusion No significantly different drug interactions were observed between fermented red ginseng and the CYP probe substrates following the two‐week administration of concentrated fermented red ginseng. However, the inhibition of P‐gp was significantly different between fermented red ginseng and the CYP probe substrates. The use of fermented red ginseng requires close attention due to the potential for increased systemic exposure when it is used in combination with P‐gp substrate drugs.
Collapse
Affiliation(s)
- Min-Gul Kim
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Yunjeong Kim
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Ji-Young Jeon
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Dal-Sik Kim
- Department of Laboratory Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| |
Collapse
|
53
|
Kim JH, Yi YS, Kim MY, Cho JY. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res 2016; 41:435-443. [PMID: 29021688 PMCID: PMC5628327 DOI: 10.1016/j.jgr.2016.08.004] [Citation(s) in RCA: 335] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/09/2016] [Indexed: 01/06/2023] Open
Abstract
Panax ginseng is one of the most universally used herbal medicines in Asian and Western countries. Most of the biological activities of ginseng are derived from its main constituents, ginsenosides. Interestingly, a number of studies have reported that ginsenosides and their metabolites/derivatives—including ginsenoside (G)-Rb1, compound K, G-Rb2, G-Rd, G-Re, G-Rg1, G-Rg3, G-Rg5, G-Rh1, G-Rh2, and G-Rp1—exert anti-inflammatory activities in inflammatory responses by suppressing the production of proinflammatory cytokines and regulating the activities of inflammatory signaling pathways, such as nuclear factor-κB and activator protein-1. This review discusses recent studies regarding molecular mechanisms by which ginsenosides play critical roles in inflammatory responses and diseases, and provides evidence showing their potential to prevent and treat inflammatory diseases.
Collapse
Affiliation(s)
- Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju, Republic of Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
54
|
Enhanced Absorption Study of Ginsenoside Compound K (20-O-β-(D-Glucopyranosyl)-20(S)-protopanaxadiol) after Oral Administration of Fermented Red Ginseng Extract (HYFRG™) in Healthy Korean Volunteers and Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3908142. [PMID: 27516803 PMCID: PMC4969531 DOI: 10.1155/2016/3908142] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/15/2016] [Indexed: 12/02/2022]
Abstract
To evaluate the pharmacokinetics of compound K after oral administration of HYFRG and RG in humans, an open-label, randomized, single-dose, fasting, and one-period pharmacokinetic study was conducted. After oral administration of a single 3 g dose of HYFRG and RG to 24 healthy Korean males, the mean (±SD) of AUC0–t and Cmax of compound K from HYFRG were 1466.83 ± 295.89 ng·h/mL and 254.45 ± 51.20 ng/mL, being 115.2- and 80-fold higher than those for RG (12.73 ± 7.83 ng·h/mL and 3.18 ± 1.70 ng/mL), respectively; in case of Sprague Dawley rats the mean (±SD) of AUC0–t and Cmax of compound K from HYFRG was 58.03 ± 32.53 ng·h/mL and 15.19 ± 10.69 ng/mL, being 6.3- and 6.0-fold higher than those from RG (9.21 ± 7.52 ng·h/mL and 2.55 ± 0.99 ng/mL), respectively. Tmax of compound K in humans and rats was 2.54 ± 0.92 and 3.33 ± 0.50 h for HYFRG and 9.11 ± 1.45 and 6.75 ± 3.97 hours for RG, respectively. In conclusion, the administration of HYFRG resulted in a higher and faster absorption of compound K in both humans and rats compared to RG.
Collapse
|
55
|
Zhou L, Zheng Y, Li Z, Bao L, Dou Y, Tang Y, Zhang J, Zhou J, Liu Y, Jia Y, Li X. Compound K Attenuates the Development of Atherosclerosis in ApoE(-/-) Mice via LXRα Activation. Int J Mol Sci 2016; 17:ijms17071054. [PMID: 27399689 PMCID: PMC4964430 DOI: 10.3390/ijms17071054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022] Open
Abstract
Background: Atherosclerosis is a fundamental pathological process responded to some serious cardiovascular events. Although the cholesterol-lowering drugs are widely prescribed for atherosclerosis therapy, it is still the leading cause of death in the developed world. Here we measured the effects of compound K in atherosclerosis formation and investigated the probably mechanisms of the anti-antherosclerosis roles of compound K. Methods: We treated the atherosclerotic model animals (apoE−/− mice on western diet) with compound K and measured the size of atherosclerotic lesions, inflammatory cytokine levels and serum lipid profile. Peritoneal macrophages were collected in vitro for the foam cell and inflammasome experiments. Results: Our results show that treatment with compound K dose-dependently attenuates the formation of atherosclerotic plaques by 55% through activation of reverse cholesterol transport pathway, reduction of systemic inflammatory cytokines and inhibition of local inflammasome activity. Compound K increases the cholesterol efflux of macrophage-derived foam cells, and reduces the inflammasome activity in cholesterol crystal stimulated macrophages. The activation of LXRα may contribute to the athero-protective effects of compound K. Conclusion: These observations provide evidence for an athero-protective effect of compound K via LXRα activation, and support its further evaluation as a potential effective modulator for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Li Zhou
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
- Department of pharmacy, Xinqiao Hospital & The Second Affiliated Hospital, Third Military Medical University, Shapingba, Chongqing 400037, China.
| | - Yu Zheng
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Zhuoying Li
- Department of Outpatient, Logistical Engineering University of PLA, Shapingba, Chongqing 401311, China.
| | - Lingxia Bao
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Yin Dou
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Yuan Tang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Jianxiang Zhang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Jianzhi Zhou
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Ya Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Yi Jia
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| |
Collapse
|
56
|
Chen J, Wang Q, Wu H, Liu K, Wu Y, Chang Y, Wei W. The ginsenoside metabolite compound K exerts its anti-inflammatory activity by downregulating memory B cell in adjuvant-induced arthritis. PHARMACEUTICAL BIOLOGY 2016; 54:1280-1288. [PMID: 27218142 DOI: 10.3109/13880209.2015.1074254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Compound K (CK, 20-O-d-glucopyranosyl-20(S)-protopanaxadiol), a novel ginsenoside metabolite, is structurally a member of the dammarane-type triterpene saponins. Several studies have identified the anti-inflammatory activity of CK. Our previous study demonstrated that CK exerted its anti-inflammatory effect via inhibition of abnormal activation and differentiation of T cells. However, its mechanism of action on B cells remains unclear. OBJECTIVE The objective of this study is to investigate the effect and underlying mechanisms of CK's effects on memory B cells in the setting of adjuvant-arthritis (AA). MATERIALS AND METHODS Complete Freund's adjuvant was used to induce AA in rats. Rats were administered, either CK (10, 40, and 160 mg/kg), once daily for 15 d, or methotrexate (MTX; 0.5 mg/kg) once every 3 d, for a total of six times. To evaluate the anti-inflammatory effect of CK, a global assessment and a swollen joint count of AA rats were performed every 3 d. Spleen index and histopathology were examined. Subsets of B cells including CD45R(+)IgM(+) (total B cells) and CD45R(+)CD27(+) (memory B cells) and expression of CD40 and CD40L were assayed by flow cytometry. RESULTS Compared with the AA rats, global assessment scores and swollen joint counts were significantly lower in the treated groups received CK (40 and 160 mg/kg; p < 0.05 and p < 0.01, respectively). CK (40 and 160 mg/kg) decreased the spleen index (p < 0.01), and alleviated hyperplasia of lymph nodes (p < 0.05 and p < 0.01, respectively) and marginal zone (p < 0.05) in the spleen. In addition, CK (40 and 160 mg/kg) suppressed memory B cell subsets (p < 0.05), and suppressed CD40L expression on T cells and CD40 expression on B cells (p < 0.05 and p < 0.01, respectively). DISCUSSION AND CONCLUSION This study demonstrated that CK downregulated memory B cells in AA rats, and this down-regulation may be T-cell dependent.
Collapse
Affiliation(s)
- Jingyu Chen
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Qingtong Wang
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Huaxun Wu
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Kangkang Liu
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Yujing Wu
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Yan Chang
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| | - Wei Wei
- a Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education , Hefei , China
| |
Collapse
|
57
|
Sun X, Jiang C, Ma L, Zhao X, Chang J, Zheng B, Li L, Xie W, Li X. 3β-Angeloyloxy-8β,10β-dihydroxyeremophila-7(11)-en-12,8α-lactone Inhibits Lipopolysaccharide-Induced Nitric Oxide Production in RAW264.7 Cells. Biol Pharm Bull 2016; 38:836-43. [PMID: 26027823 DOI: 10.1248/bpb.b14-00709] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Farfugium japonicum (L.) KITAM, named "Lian-Peng-Cao" in China, has been traditionally used in Chinese folk medicine to treat sore throat, cold and cough due to its anti-inflammatory properties. In this study, the anti-inflammatory action of 3β-angeloyloxy-8β,10β-dihydroxyeremophila-7(11)-en-12,8α-lactone (FJ1) isolated from Farfugium japonicum and its molecular mechanism in RAW264.7 cells were investigated. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed that FJ1 with or without 3 µg/mL lipopolysaccharide (LPS) had no significant cytotoxicity in RAW264.7 cells. The production of nitric oxide (NO) was identified with a Griess reagent kit. The mRNA expression of inducible nitric oxide synthase (iNOS) and cytokines, including tumor necrosis factor α (TNF-α) and cyclooxygenase-2 (COX-2), was measured by real-time polymerase chain reaction (PCR). Reactive oxygen species (ROS) production was detected by flow cytometry analysis. Western blot was used to examine the protein expression of nuclear factor-kappa B (NF-κB)/p65, inhibitor of kappa B (IκB)-α, phosphorylated IκB-α (p-IκB-α), mitogen-activated protein kinase (MAPK) molecules, iNOS, and TNF-α. We discovered that FJ1 possesses anti-inflammatory effects that inhibit the release of LPS-stimulated pro-inflammatory cytokines, including NO and ROS. The molecular mechanism of FJ1-mediated anti-inflammation is associated with decreasing phosphorylation of MAPK molecules, including extracellular signal-related kinase 1/2 (ERK1/2), p38 MAPK, and c-Jun NH2-terminal kinase (JNK), FJ1 also reverses IκB degradation and attenuates the mRNA and protein expression of NF-κB-related downstream inducible enzymes and cytokines, such as iNOS, TNF-α in RAW264.7 cells. The results suggest that FJ1 has anti-inflammatory properties, which indicates that F. japonicum can be utilized to treat inflammatory diseases. The potential mechanism is associated with the NF-κB and MAPK activation pathways in LPS-stimulated macrophages.
Collapse
Affiliation(s)
- Xiao Sun
- School of Ocean, Shandong University
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Microbial conversion of major ginsenosides in ginseng total saponins by Platycodon grandiflorum endophytes. J Ginseng Res 2015; 40:366-374. [PMID: 27746689 PMCID: PMC5052436 DOI: 10.1016/j.jgr.2015.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 11/14/2015] [Accepted: 11/27/2015] [Indexed: 01/31/2023] Open
Abstract
Background In this study, we screened and identified an endophyte JG09 having strong biocatalytic activity for ginsenosides from Platycodon grandiflorum, converted ginseng total saponins and ginsenoside monomers, determined the source of minor ginsenosides and the transformation pathways, and calculated the maximum production of minor ginsenosides for the conversion of ginsenoside Rb1 to assess the transformation activity of endophyte JG09. Methods The transformation of ginseng total saponins and ginsenoside monomers Rb1, Rb2, Rc, Rd, Rg1 into minor ginsenosides F2, C-K and Rh1 using endophyte JG09 isolated by an organizational separation method and Esculin-R2A agar assay, as well as the identification of transformed products via TLC and HPLC, were evaluated. Endophyte JG09 was identified through DNA sequencing and phylogenetic analysis. Results A total of 32 β-glucosidase-producing endophytes were screened out among the isolated 69 endophytes from P. grandiflorum. An endophyte bacteria JG09 identified as Luteibacter sp. effectively converted protopanaxadiol-type ginsenosides Rb1, Rb2, Rc, Rd into minor ginsenosides F2 and C-K, and converted protopanaxatriol-type ginsenoside Rg1 into minor ginsenoside Rh1. The transformation pathways of major ginsenosides by endophyte JG09 were as follows: Rb1→Rd→F2→C-K; Rb2→C-O→C-Y→C-K; Rc→C-Mc1→C-Mc→C-K; Rg1→Rh1. The maximum production rate of ginsenosides F2 and C-K reached 94.53% and 66.34%, respectively. Conclusion This is the first report about conversion of major ginsenosides into minor ginsenosides by fermentation with P. grandiflorum endophytes. The results of the study indicate endophyte JG09 would be a potential microbial source for obtaining minor ginsenosides.
Collapse
|
59
|
Oh HA, Kim DE, Choi HJ, Kim NJ, Kim DH. Anti-stress effects of 20(S)-protopanaxadiol and 20(S)-protopanaxatriol in immobilized mice. Biol Pharm Bull 2015; 38:331-5. [PMID: 25747994 DOI: 10.1248/bpb.b14-00669] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Panax ginseng C.A. MEYER (Araliaceae), which contains ginsenosides as its main components, has been shown to have various biological effects, including anti-inflammatory, anxiolytic, anti-stress, and anti-tumor effects. Orally administered ginsenoside Rb1 and Re are metabolized to 20(S)-protopanaxadiol (PPD) and compound K via ginsenoside Rd and 20(S)-protopanaxatriol (PPT) and ginsenoside Rh1 via ginsenoside Rg1 by gut microbiota, respectively. Therefore, we investigated the anti-stress effects of these metabolites, PPD and PPT, by measuring their anxiolytic and anti-inflammatory effects in immobilized mice. Treatment with PPD and PPT prior to immobilization stress increased the time spent in open arms and open arm entries in the elevated plus-maze (EPM) test. The anxiolytic effects of PPD (10 mg/kg) and PPT (10 mg/kg) were comparable to that of buspirone (1 mg/kg). This observed anxiolytic effect of PPD was significantly blocked by flumazenil or bicuculline, and the effect of PPT was blocked by WAY-100635. Treatment with PPD also potently suppressed immobilization stress-induced serum levels of corticosterone and interleukin (IL)-6 by the enzyme-linked immunosorbent assay. However, PPT treatment did not suppress them. Based on these findings, PPD and PPT may exhibit the anxiolytic effect via γ-aminobutyrateA (GABAA) receptor(s) and serotonergic receptor(s), respectively, and PPD may have an anti-inflammatory effect that is more potent than that of PPT.
Collapse
Affiliation(s)
- Hyun A Oh
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University
| | | | | | | | | |
Collapse
|
60
|
Chen W, Wang J, Luo Y, Wang T, Li X, Li A, Li J, Liu K, Liu B. Ginsenoside Rb1 and compound K improve insulin signaling and inhibit ER stress-associated NLRP3 inflammasome activation in adipose tissue. J Ginseng Res 2015; 40:351-358. [PMID: 27746687 PMCID: PMC5052408 DOI: 10.1016/j.jgr.2015.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/13/2015] [Accepted: 11/18/2015] [Indexed: 12/18/2022] Open
Abstract
Background This study was designed to investigate whether ginsenoside Rb1 (Rb1) and compound K (CK) ameliorated insulin resistance by suppressing endoplasmic reticulum (ER) stress-induced inflammation in adipose tissue. Methods To induce ER stress, epididymal adipose tissue from mice or differentiated 3T3 adipocytes were exposed to high glucose. The effects of Rb1 and CK on reactive oxygen species production, ER stress, TXNIP/NLRP3 inflammasome activation, inflammation, insulin signaling activation, and glucose uptake were detected by western blot, emzyme-linked immunosorbent assay, or fluorometry. Results Rb1 and CK suppressed ER stress by dephosphorylation of IRE1α and PERK, thereby reducing TXNIP-associated NLRP3 inflammasome activation in adipose tissue. As a result, Rb1 and CK inhibited IL-1β maturation and downstream inflammatory factor IL-6 secretion. Inflammatory molecules induced insulin resistance by upregulating phosphorylation of insulin receptor substrate-1 at serine residues and impairing insulin PI3K/Akt signaling, leading to decreased glucose uptake by adipocytes. Rb1 and CK reversed these changes by inhibiting ER stress-induced inflammation and ameliorating insulin resistance, thereby improving the insulin IRS-1/PI3K/Akt-signaling pathway in adipose tissue. Conclusion Rb1 and CK inhibited inflammation and improved insulin signaling in adipose tissue by suppressing ER stress-associated NLRP3 inflammation activation. These findings offered novel insight into the mechanism by which Rb1 and CK ameliorate insulin resistance in adipose tissue.
Collapse
Affiliation(s)
- Weijie Chen
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Junlian Wang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Yong Luo
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Tao Wang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Xiaochun Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Aiyun Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Jia Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Kang Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Baolin Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
61
|
Ginsenoside Rd Improves Learning and Memory Ability in APP Transgenic Mice. J Mol Neurosci 2015; 57:522-8. [PMID: 26358038 DOI: 10.1007/s12031-015-0632-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease (AD) is a complicated neurodegenerative disease which causes memory loss and dementia. Many researchers have revealed the vital roles of β-amyloid proteins (Aβ) in the proceeds of AD. Aβ deposition in AD patients' brains might function as immune stimulus, and inflammation is believed to play an important role in AD pathologically. We experimentally used amyloid β-protein precursor (APP) transgenic (Tg) mice in this study to further clarify the neuroprotective effects of ginsenoside Rd on AD and its possible mechanisms. It was found that Rd could improve learning and memory ability in APP Tg mice, probably through inhibiting the transcription activity of NFκB. With the activation of the NFκB pathway being suppressed, the reduction of pro-inflammatory cytokines and the generation of protective factors had been increased ultimately. In conclusion, Rd had a neuroprotective effect on APP Tg mice, and it can be used as an alternative drug therapy in AD patients for their memory dysfunction.
Collapse
|
62
|
Tsai TC, Chen HY, Sheu JH, Chiang MY, Wen ZH, Dai CF, Su JH. Structural Elucidation and Structure-Anti-inflammatory Activity Relationships of Cembranoids from Cultured Soft Corals Sinularia sandensis and Sinularia flexibilis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7211-7218. [PMID: 26260702 DOI: 10.1021/acs.jafc.5b01931] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
New cembranoids 4-carbomethoxyl-10-epigyrosanoldie E (1), 7-acetylsinumaximol B (2), diepoxycembrene B (6), dihydromanaarenolide I (8), and isosinulaflexiolide K (9), along with 11 known related metabolites, were isolated from cultured soft corals Sinularia sandensis and Sinularia flexibilis. The structures were elucidated by means of infrared, mass spectrometry, and nuclear magnetic resonance techniques, and the absolute configurations of 1, 4, 9, and 15 were further confirmed by single-crystal X-ray diffraction analysis. The absolute configurations of these coral metabolites and comparison with known analogues showed that one hypothesis (that cembrane diterpenes possessing an absolute configuration of an isopropyl group at C1 obtained from Alcyonacean soft corals belong to the α series, whereas analogues isolated from Gorgonacean corals belong to the β series) is not applicable for a small number of cembranoids. An in vitro anti-inflammatory study using LPS-stimulated macrophage-like cell line RAW 264.7 revealed that compounds 9-14 significantly suppressed the accumulation of pro-inflammatory proteins, iNOS and COX-2. Structure-activity relationship analysis indicated that cembrane-type compounds with one seven-membered lactone moiety at C-1 are potential anti-inflammatory agents. This is the first culture system in the world that has successfully been used to farm S. sandensis.
Collapse
Affiliation(s)
- Tsung-Chang Tsai
- †Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- ‡Department of Beauty Science, Meiho University, Pingtung 912, Taiwan
| | - Hsueh-Yu Chen
- ∥Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Jyh-Horng Sheu
- †Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Michael Y Chiang
- ⊥Department of Chemistry, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Zhi-Hong Wen
- †Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chang-Feng Dai
- #Institute of Oceanography, National Taiwan University, Taipei 106, Taiwan
| | - Jui-Hsin Su
- ∥Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| |
Collapse
|
63
|
Lee SY, Jeong JJ, Eun SH, Kim DH. Anti-inflammatory effects of ginsenoside Rg1 and its metabolites ginsenoside Rh1 and 20(S)-protopanaxatriol in mice with TNBS-induced colitis. Eur J Pharmacol 2015; 762:333-43. [PMID: 26054809 DOI: 10.1016/j.ejphar.2015.06.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/26/2015] [Accepted: 06/04/2015] [Indexed: 10/23/2022]
Abstract
Ginsenoside Rg1, one of the main constituents of Panax ginseng, exhibits anti-inflammatory effect. In a preliminary study, it was observed that ginsenoside Rg1 was metabolized to 20(S)-protopanaxtriol via ginsenosides Rh1 and F1 by gut microbiota. We further investigated the anti-inflammatory effects of ginsenoside Rg1 and its metabolites in vitro and in vivo. Ginsenosides Rg1, Rh1, and 20(S)-protopanaxtriol inhibited the activation of NF-κB activation, phosphorylation of transforming growth factor beta-activated kinase 1 and interleukin (IL)-1 receptor-associated kinase, and expression of tumor necrosis factor-α and IL-1β in lipopolysaccharide (LPS)-stimulated macrophages. They also inhibited the binding of LPS to toll-like receptor 4 on the macrophages. Orally administered ginsenoside Rg1, Rh1, or 20(S)-protopanaxtriol inhibited 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colon shortening, myeloperoxidase activity, and expression of IL-1β, IL-17, and tumor necrosis factor-α in mice with TNBS-induced colitis. They did not only inhibit TNBS-induced NF-κB activation, but also restored TNBS-induced Th17/Treg imbalance. They restored IL-10 and Foxp3 expression. Moreover, they inhibited Th17 cell differentiation in vitro. Of these metabolites, in vitro and in vivo anti-inflammatory effect of 20(S)-protopanaxtriol was the most potent, followed by Rh1. These findings suggest that ginsenoside Rg1 is metabolized to 20(S)-protopanaxtriol via ginsenosides Rh1 and F1 and these metabolites particularly 20(S)-protopanaxtriol, may ameliorate inflammatory disease such as colitis by inhibiting the binding of LPS to TLR4 on macrophages and restoring the Th17/Treg imbalance.
Collapse
Affiliation(s)
- Sang-Yun Lee
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Su-Hyeon Eun
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1, Hoegi, Dongdaemun-ku, Seoul 130-701, Republic of Korea.
| |
Collapse
|
64
|
Isolation and identification of antiproliferative substances from ginseng fermented using Ganoderma lucidum mycelia. Food Sci Biotechnol 2015. [DOI: 10.1007/s10068-015-0074-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
65
|
Chen J, Wu H, Wang Q, Chang Y, Liu K, Wei W. Ginsenoside metabolite compound K suppresses T-cell priming via modulation of dendritic cell trafficking and costimulatory signals, resulting in alleviation of collagen-induced arthritis. J Pharmacol Exp Ther 2015; 353:71-9. [PMID: 25630466 DOI: 10.1124/jpet.114.220665] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ginsenoside metabolite compound K (CK; 20-O-d-glucopyranosyl-20(S)-protopanaxadiol), a novel ginsenoside metabolite, belongs to the dammarane-type triterpene saponins, according to its structure. The anti-inflammatory activity of CK has been identified in several studies. Our study demonstrated that CK exerted an anti-inflammatory effect in collagen-induced arthritis (CIA) and adjuvant-induced arthritis animal models, and this effect was due to inhibition of the abnormal activation and differentiation of T cells. However, the mechanism of CK in suppressing T-cell activation remains unclear. In this study, CK had a therapeutic effect in mice with CIA, decreased the percentage of activated T cells and dendritic cells (DCs), and increased the percentage of naive T cells in lymph nodes. The inhibitory effect on T-cell activation of CK was related to suppression of accumulation of DCs in lymph nodes. CK decreased CCL21 levels in lymph nodes and CCR7 expression in DCs and suppressed CCL21/CCR7-mediated migration of DCs, thus reducing accumulation of DCs in lymph nodes. In addition, signals for T-cell activation including major histocompatibility complex class II and costimulatory molecules, such as CD80 and CD86, were suppressed by CK, and the proliferation of T cells induced by DCs was inhibited by CK. In conclusion, this study demonstrated that CK downregulated DC priming of T-cell activation in CIA, and suppression of CCL21/CCR7-mediated DC migration and signaling between T cells and DCs might be the potential mechanism. These results provide an interesting, novel insight into the potential mechanism by which CK contributes to the anti-inflammatory effect in autoimmune conditions.
Collapse
Affiliation(s)
- Jingyu Chen
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Yan Chang
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Kangkang Liu
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology of Anhui Medical University and Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| |
Collapse
|
66
|
Yang XD, Yang YY, Ouyang DS, Yang GP. A review of biotransformation and pharmacology of ginsenoside compound K. Fitoterapia 2015; 100:208-20. [DOI: 10.1016/j.fitote.2014.11.019] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 11/19/2014] [Accepted: 11/21/2014] [Indexed: 12/14/2022]
|
67
|
Xu R, Peng Y, Wang M, Fan L, Li X. Effects of broad-spectrum antibiotics on the metabolism and pharmacokinetics of ginsenoside Rb1: a study on rats׳ gut microflora influenced by lincomycin. JOURNAL OF ETHNOPHARMACOLOGY 2014; 158 Pt A:338-344. [PMID: 25446586 DOI: 10.1016/j.jep.2014.10.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 06/04/2023]
Abstract
Ginsenoside Rb1 is a biologically active compound that is abundant in ginseng (Panax ginseng). It has been reported that ginsenosides could be metabolized by enzymes and bacteria in the large intestine. In this study, the effects of intestinal bacteria on the metabolism and pharmacokinetics of ginsenoside Rb1 were investigated using lincomycin-treated rat models (4.8g/kg and 0.12g/kg). Specifically, ginsenoside Rb1 was incubated anaerobically with rat fecal suspensions obtained from the control and two model groups at 0, 6, 12, 24, and 48h. Ginsenoside Rb1 and its metabolites were determined by HPLC analysis. Compared with the normal rats case where Rd and compound K were detected in the incubation mixture, ginsenoside Rd and F2 were found in the 0.12g/kg group, but only Rd was found in the 4.8g/kg group. Moreover, fecal β-glucosidase activity was significantly lower in lincomycin-treated (0.12g/kg and 4.8g/kg) model rats. After administration of Rb1 to rats, ginsenoside Rb1 and its metabolites Rd, Rg3, and Rh2 were detectable in normal rat urine, whereas none was detected in the two model groups. The plasma concentration-time Rb1 were compared between model groups and normal rats. The systemic exposure as evidenced by the AUC and T1/2 values was significantly higher in model groups than in normal rats. Our findings demonstrated that consumption of lincomycin could lead to the formation of specific metabolites and pharmacokinetic changes of ginsenoside Rb1 in the gut, attributed to alterations in metabolic activities of intestinal bacteria. Our results also suggested that patients who want to use intestinal bacteria-metabolized drugs such as ginseng (Panax ginseng) should pay attention to the profile of intestinal bacteria or potential drug interactions to ensure therapeutic efficacy.
Collapse
Affiliation(s)
- Renjie Xu
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Ying Peng
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Mengyue Wang
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Lirong Fan
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China.
| |
Collapse
|
68
|
Hong SM, Jeon SO, Seo JE, Chun KH, Oh DH, Choi YW, Lee DI, Jeong SH, Kang JS, Lee S. Fabrication of Compound K-loaded Polymeric Micelle System and its Characterization in vitro and Oral Absorption Enhancement in vivo. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.11.3188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
69
|
Fang HY, Chiou SF, Uvarani C, Wen ZH, Hsu CH, Wu YC, Wang WL, Liaw CC, Sheu JH. Cytotoxic, Anti-inflammatory, and Antibacterial Sulfur-Containing Polybromoindoles from the Formosan Red Alga Laurencia brongniartii. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2014. [DOI: 10.1246/bcsj.20140165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Hui-Yu Fang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Shu-Fen Chiou
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Chokkalingam Uvarani
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Zih-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Chi-Hsin Hsu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Yang-Chang Wu
- Center for Molecular Medicine, China Medical University Hospital, and School of Pharmacy, China Medical University
| | - Wei-Lung Wang
- Department of Biology, National Changhua University of Education
| | - Chih-Chuang Liaw
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University
- Asia-Pacific Ocean Research Center, National Sun Yat-sen University
- Graduate Institute of Natural Products, Kaohsiung Medical University
- Department of Medical Research, China Medical University Hospital, China Medical University
| |
Collapse
|
70
|
Hou YL, Tsai YH, Lin YH, Chao JCJ. Ginseng extract and ginsenoside Rb1 attenuate carbon tetrachloride-induced liver fibrosis in rats. Altern Ther Health Med 2014; 14:415. [PMID: 25344394 PMCID: PMC4216840 DOI: 10.1186/1472-6882-14-415] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023]
Abstract
Background Ginsenosides, the major bioactive compounds in ginseng root, have been found to have antioxidant, immunomodulatory and anti-inflammatory activities. This study investigated the effects of ginsenosides on carbon tetrachloride (CCl4)-induced hepatitis and liver fibrosis in rats. Methods Male Sprague–Dawley rats were randomly divided into four groups: control, CCl4, CCl4 + 0.5 g/kg Panax ginseng extract and CCl4 + 0.05 g/kg ginsenoside Rb1 groups. The treated groups were orally given Panax ginseng extract or ginsenoside Rb1 two weeks before the induction of liver injury for successive 9 weeks. Liver injury was induced by intraperitoneally injected with 400 ml/l CCl4 at a dose of 0.75 ml/kg body weight weekly for 7 weeks. The control group was intraperitoneally injected with olive oil. Results The pathological results showed that ginsenoside Rb1 decreased hepatic fat deposition (2.65 ± 0.82 vs 3.50 ± 0.75, p <0.05) and Panax ginseng extract lowered hepatic reticular fiber accumulation (1.05 ± 0.44 vs 1.60 ± 0.39, p <0.01) increased by CCl4. Plasma alanine aminotransferase and aspartate aminotransferase activities were increased by CCl4 (p <0.01), and aspartate aminotransferase activity was decreased by Panax ginseng extract at week 9 (p <0.05). Exposure to CCl4 for 7 weeks, the levels of plasma and hepatic triglycerides (p <0.01), hepatic cholesterol (p <0.01), interleukin-1β (p <0.01), prostaglandin E2 (p <0.05), soluble intercellular adhesion molecule-1 (p <0.05), hydroxyproline (p <0.05), matrix metalloproteinase-2 (p <0.05) and tissue inhibitor of metalloproteinase-1 (TIMP-1) (p <0.01) were elevated, however, hepatic interleukin-10 level was lowered (p <0.05). Both Panax ginseng extract and ginsenoside Rb1 decreased plasma and hepatic triglyceride, hepatic prostaglandin E2, hydroxyproline and TIMP-1 levels, and Panax ginseng extract further inhibited interleukin-1β concentrations (p <0.05). Conclusions Panax ginseng extract and ginsenoside Rb1 attenuate plasma aminotransferase activities and liver inflammation to inhibit CCl4-induced liver fibrosis through down-regulation of hepatic prostaglandin E2 and TIMP-1.
Collapse
|
71
|
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: 45] [Impact Index Per Article: 4.5] [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
|
72
|
Song YS, Lee Y, Kwon TR, Kim YH, Kim BJ. Picrasma quassioides inhibits LPS- and IFN-γ-stimulated nitric oxide production and inflammatory response in RAW264.7 macrophage cells. BIOTECHNOL BIOPROC E 2014. [DOI: 10.1007/s12257-014-0131-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
73
|
Samimi R, Salarian M, Xu WZ, Lui EMK, Charpentier PA. Encapsulation of Acetyl Ginsenoside Rb1 within Monodisperse Poly(dl-lactide-co-glycolide) Microspheres Using a Microfluidic Device. Ind Eng Chem Res 2014. [DOI: 10.1021/ie501118u] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Raziye Samimi
- Department of Chemical and Biochemical Engineering, ‡Biomedical Engineering Graduate Program, §Department of Physiology and Pharmacology, and ∥The Ontario Ginseng Innovation & Research Consortium, University of Western Ontario, London, Ontario, Canada, N6A 5B9
| | - Mehrnaz Salarian
- Department of Chemical and Biochemical Engineering, ‡Biomedical Engineering Graduate Program, §Department of Physiology and Pharmacology, and ∥The Ontario Ginseng Innovation & Research Consortium, University of Western Ontario, London, Ontario, Canada, N6A 5B9
| | - William Z. Xu
- Department of Chemical and Biochemical Engineering, ‡Biomedical Engineering Graduate Program, §Department of Physiology and Pharmacology, and ∥The Ontario Ginseng Innovation & Research Consortium, University of Western Ontario, London, Ontario, Canada, N6A 5B9
| | - Edmund M. K. Lui
- Department of Chemical and Biochemical Engineering, ‡Biomedical Engineering Graduate Program, §Department of Physiology and Pharmacology, and ∥The Ontario Ginseng Innovation & Research Consortium, University of Western Ontario, London, Ontario, Canada, N6A 5B9
| | - Paul A. Charpentier
- Department of Chemical and Biochemical Engineering, ‡Biomedical Engineering Graduate Program, §Department of Physiology and Pharmacology, and ∥The Ontario Ginseng Innovation & Research Consortium, University of Western Ontario, London, Ontario, Canada, N6A 5B9
| |
Collapse
|
74
|
Ginsenoside Metabolite Compound K Alleviates Adjuvant-Induced Arthritis by Suppressing T Cell Activation. Inflammation 2014; 37:1608-15. [DOI: 10.1007/s10753-014-9887-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
75
|
Ginsenoside compound K inhibits angiogenesis via regulation of sphingosine kinase-1 in human umbilical vein endothelial cells. Arch Pharm Res 2014; 37:1183-92. [PMID: 24687256 DOI: 10.1007/s12272-014-0340-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
Abstract
Ginsenoside compound K (CK) is a metabolite of the protopanaxadiol-type saponins of Panax ginseng C.A. Meyer (Araliaceae), has long been used to treat against the development of cancer, inflammation, allergies, and diabetes. This study examined the anti-angiogenic properties of CK against sphingosine 1-phosphate (S1P)-induced cell migration via regulation of sphingosine kinase 1 (SPHK1) in human umbilical vein endothelial cells (HUVEC). Studies on S1P-induced cell migration, expression of SPHK1 and MMPs and analysis of sphingolipid metabolites by LC-MS/MS were examined after the treatment of CK (2.5, 5, 10 μg/mL) in HUVEC. S1P produced by SPHK1 is also involved in cell growth, migration, and protection of apoptosis; therefore, we sought to investigate whether ginsenosides are able to regulate SPHK1. For this purpose, we developed an inhibitory assay of SPHK1 activity and an analytical method for detection of S1P and other sphingolipid metabolites in HUVEC. Ginsenoside CK inhibited 100 nM S1P-induced cell migrations in a dose-dependent manner. Among tested ginsenosides, CK exclusively inhibited S1P production, SPHK1 activity and SPHK1 expression in HUVEC, whereas expression of the pro-apoptotic sphingolipids, sphingosine and ceramide, was increased in response to CK. The major subspecies of the increased ceramide was C24:0-ceramide. CK also disrupted the sphingolipid rheostat, which ultimately influences cell fate, and dose-dependently inhibited HUVEC migration by reducing expression of metalloproteinases (MMPs). Ginsenoside CK acts as a unique HUVEC migration inhibitor by regulating MMP expression, as well as the activity of SPHK1 and its related sphingolipid metabolites.
Collapse
|
76
|
Smith I, Williamson EM, Putnam S, Farrimond J, Whalley BJ. Effects and mechanisms of ginseng and ginsenosides on cognition. Nutr Rev 2014; 72:319-33. [DOI: 10.1111/nure.12099] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Imogen Smith
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | - Elizabeth M Williamson
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | | | | | - Benjamin J Whalley
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| |
Collapse
|
77
|
Choi HS, Seo HS, Kim SR, Choi YK, Shin YC, Ko SG. Anti-inflammatory and anti-proliferative effect of herbal medicines (APR) in RAW264.7 cells. Mol Med Rep 2014; 9:1569-74. [PMID: 24626965 PMCID: PMC4020494 DOI: 10.3892/mmr.2014.2033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 02/24/2014] [Indexed: 12/31/2022] Open
Abstract
The objective of the present study was to analyze the effect of a mixture of medicinal plants [Angelica gigas Nakai, Panax ginseng and Rhus verniciflua Stokes (APR)] on lipopolysaccharide (LPS)-induced inflammatory responses in the murine macrophage cell line RAW264.7. Cells were treated with APR and LPS at various concentrations and indicated times. WST assay, trypan blue assay and quantification of activated cells demonstrated that APR suppressed cell proliferation in a dose-dependent manner. APR induced G1 cell cycle arrest and inhibited the LPS-induced phosphorylation of protein kinase B (AKT), extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (p38) and necrosis factor κB (NF-κB). APR also suppressed nitric oxide synthase isoform (iNOS) and prostaglandin endoperoxide synthase 2 (Cox-2) messenger ribonucleic acid (mRNA) expression induced by LPS. Furthermore, APR decreased LPS-induced intracellular reactive oxygen species (ROS) levels, mitochondrial membrane potential, as well as induced PARP and caspase-3 cleavage, suggesting that APR causes apoptosis. In conclusion, the present study indicated that APR may be advantageous in treating inflammatory disease.
Collapse
Affiliation(s)
- Han-Seok Choi
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Hye Sook Seo
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Soon Re Kim
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Youn Kyung Choi
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Yong-Cheol Shin
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Seong-Gyu Ko
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, Institute of Oriental Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| |
Collapse
|
78
|
Samimi R, Xu WZ, Alsharari Q, Charpentier PA. Supercritical fluid chromatography of North American ginseng extract. J Supercrit Fluids 2014. [DOI: 10.1016/j.supflu.2013.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
79
|
Choi JS, Chun KS, Kundu J, Kundu JK. Biochemical basis of cancer chemoprevention and/or chemotherapy with ginsenosides (Review). Int J Mol Med 2013; 32:1227-38. [PMID: 24126942 DOI: 10.3892/ijmm.2013.1519] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/26/2013] [Indexed: 11/06/2022] Open
Abstract
Cancer still imposes a global threat to public health. After decades of research on cancer biology and enormous efforts in developing anticancer therapies, we now understand that the majority of cancers can be prevented. Bioactive phytochemicals present in edible plants have been shown to reduce the risk of various types of cancer. Ginseng (Panax ginseng C.A. Meyer), which contains a wide variety of saponins, known as ginsenosides, is an age-old remedy for human ailments, including cancer. Numerous laboratory-based studies have revealed the anticancer properties of ginsenosides, which compel tumor cells to commit suicide, arrest the proliferation of cancer cells in culture and inhibit experimentally-induced tumor formation in laboratory animals. Ginsenosides have been reported to inhibit tumor angiogenesis, as well as the invasion and metastasis of various types of cancer cells. Moreover, ginsenosides as combination therapy enhance the sensitivity of chemoresistant tumors to clinically used chemotherapeutic agents. This review sheds light on the molecular mechanisms underlying the cancer chemopreventive and/or chemotherapeutic activity of ginsenosides and their intestinal metabolites with particular focus on the modulation of cell signaling pathways associated with oxidative stress, inflammation, cell proliferation, apoptosis, angiogenesis and the metastasis of cancer cells.
Collapse
Affiliation(s)
- Joon-Seok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | |
Collapse
|
80
|
Shen T, Lee J, Park MH, Lee YG, Rho HS, Kwak YS, Rhee MH, Park YC, Cho JY. Ginsenoside Rp1, a Ginsenoside Derivative, Blocks Promoter Activation of iNOS and COX-2 Genes by Suppression of an IKKβ-mediated NF-кB Pathway in HEK293 Cells. J Ginseng Res 2013; 35:200-8. [PMID: 23717062 PMCID: PMC3659523 DOI: 10.5142/jgr.2011.35.2.200] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 03/29/2011] [Accepted: 03/30/2011] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside (G) Rp1 is a ginseng saponin derivative with anti-cancer and anti-inflammatory activities. In this study, we examined the mechanism by which G-Rp1 inhibits inflammatory responses of cells. We did this using a strategy in which DNA constructs containing cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) promoters were transfected into HEK293 cells. G-Rp1 strongly inhibited the promoter activities of COX-2 and iNOS; it also inhibited lipopolysaccharide induced upregulation of COX-2 and iNOS mRNA levels in RAW264.7 cells. In HEK293 cells G-Rp1 did not suppress TANK binding kinase 1-, Toll-interleukin-1 receptor-domain-containing adapter-inducing interferon-β (TRIF)-, TRIFrelated adaptor molecule (TRAM)-, or activation of interferon regulatory factor (IRF)-3 and nuclear factor (NF)-кB by the myeloid differentiation primary response gene (MyD88)-induced. However, G-Rp1 strongly suppressed NF-кB activation induced by IкB kinase (IKK)β in HEK293 cells. Consistent with these results, G-Rp1 substantially inhibited IKKβ-induced phosphorylation of IкBɑ and p65. These results suggest that G-Rp1 is a novel anti-inflammatory ginsenoside analog that can be used to treat IKKβ/NF-кB-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Ting Shen
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Yuan HD, Quan HY, Jung MS, Kim SJ, Huang B, Kim DY, Chung SH. Anti-Diabetic Effect of Pectinase-Processed Ginseng Radix (GINST) in High Fat Diet-Fed ICR Mice. J Ginseng Res 2013; 35:308-14. [PMID: 23717074 PMCID: PMC3659543 DOI: 10.5142/jgr.2011.35.3.308] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/21/2011] [Accepted: 05/21/2011] [Indexed: 11/18/2022] Open
Abstract
In the present study, we investigate anti-diabetic effect of pectinase-processed ginseng radix (GINST) in high fat diet-fed ICR mice. The ICR mice were divided into three groups: regular diet group, high fat diet control group (HFD), and GINST-treated group. To induce hyperglycemia, mice were fed a high fat diet for 10 weeks, and mice were administered with 300 mg/ kg of GINST once a day for 5 weeks. Oral glucose tolerance test revealed that GINST improved glucose tolerance after glucose challenge. Compared to the HFD control group, fasting blood glucose and insulin levels were decreased by 57.8% (p<0.05) and 30.9% (p<0.01) in GINST-treated group, respectively. With decreased plasma glucose and insulin levels, the insulin resistance index of the GINST-treated group was reduced by 68.1% (p<0.01) compared to the HFD control group. Pancreas of GINST-treated mice preserved a morphological integrity of islets and consequently having more insulin contents. In addition, GINST up-regulated the levels of phosphorylated AMP-activated protein kinase (AMPK) and its target molecule, glucose transporter 4 (GLUT4) protein expression in the skeletal muscle. Our results suggest that GINST ameliorates a hyperglycemia through activation of AMPK/ GLUT4 signaling pathway, and has a therapeutic potential for type 2 diabetes.
Collapse
Affiliation(s)
- Hai Dan Yuan
- Department of Life and Nanopharmaceutical Science, Graduate School of Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | |
Collapse
|
82
|
Kim DH. Chemical Diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng. J Ginseng Res 2013; 36:1-15. [PMID: 23717099 PMCID: PMC3659563 DOI: 10.5142/jgr.2012.36.1.1] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 10/31/2011] [Accepted: 10/31/2011] [Indexed: 11/18/2022] Open
Abstract
The major commercial ginsengs are Panax ginseng Meyer (Korean ginseng), P. quinquifolium L. (American ginseng), and P. notoginseng (Burk.) FH Chen (Notoginseng). P. ginseng is the most commonly used as an adaptogenic agent and has been shown to enhance physical performance, promote vitality, increase resistance to stress and aging, and have immunomodulatory activity. These ginsengs contain saponins, which can be classified as dammarane-type, ocotillol-type and oleanane-type oligoglycosides, and polysaccharides as main constituents. Dammarane ginsenosides are transformed into compounds such as the ginsenosides Rg3, Rg5, and Rk1 by steaming and heating and are metabolized into metabolites such as compound K, ginsenoside Rh1, protoand panaxatriol by intestinal microflora. These metabolites are nonpolar, pharmacologically active and easily absorbed from the gastrointestinal tract. However, the activities metabolizing these constituents into bioactive compounds differ significantly among individuals because all individuals possess characteristic indigenous strains of intestinal bacteria. To overcome this difference, ginsengs fermented with enzymes or microbes have been developed.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmaceutical Science, Kyung Hee University, Seoul 130-701, Korea
| |
Collapse
|
83
|
Kim S, Na JY, Song KB, Choi DS, Kim JH, Kwon YB, Kwon J. Protective Effect of Ginsenoside Rb1 on Hydrogen Peroxide-induced Oxidative Stress in Rat Articular Chondrocytes. J Ginseng Res 2013; 36:161-8. [PMID: 23717116 PMCID: PMC3659580 DOI: 10.5142/jgr.2012.36.2.161] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 02/10/2012] [Accepted: 02/10/2012] [Indexed: 11/23/2022] Open
Abstract
The abnormal maturation and ossification of articular chondrocytes play a central role in the pathogenesis of osteoarthritis (OA). Inhibiting the enzymatic degradation of the extracellular matrix and maintaining the cellular phenotype are two of the major goals of interest in managing OA. Ginseng is frequently taken orally, as a crude substance, as a traditional medicine in Asian countries. Ginsenoside Rb1, a major component of ginseng that contains an aglycone with a dammarane skeleton, has been reported to exhibit various biological activities, including anti-inflammatory and anti-tumor effects. However, a chondroprotective effect of ginsenoside Rb1 related to OA has not yet been reported. The purpose of this study was to demonstrate the chondroprotective effect of ginsenoside Rb1 on the regulation of pro-inflammatory factors and chondrogenic genes. Cultured rat articular chondrocytes were treated with 100 μM ginsenoside Rb1 and/or 500 μM hydrogen peroxide (H2O2) and assessed for viability, reactive oxygen species production, nitric oxide (NO) release, and chondrogenic gene expression. Ginsenoside Rb1 treatment resulted in reductions in the levels of pro-inflammatory cytokine and NO in H2O2-treated chondrocytes. The expression levels of chondrogenic genes, such as type II collagen and SOX9, were increased in the presence of ginsenoside Rb1, whereas the expression levels of inflammatory genes related to chondrocytes, such as MMP1 and MMP13, were reduced by approximately 50%. These results suggest that ginsenoside Rb1 has potential for use as a therapeutic agent in OA patients.
Collapse
Affiliation(s)
- Sokho Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-756, Korea
| | | | | | | | | | | | | |
Collapse
|
84
|
Kim DH, Chung JH, Yoon JS, Ha YM, Bae S, Lee EK, Jung KJ, Kim MS, Kim YJ, Kim MK, Chung HY. Ginsenoside Rd inhibits the expressions of iNOS and COX-2 by suppressing NF-κB in LPS-stimulated RAW264.7 cells and mouse liver. J Ginseng Res 2013; 37:54-63. [PMID: 23717157 PMCID: PMC3659628 DOI: 10.5142/jgr.2013.37.54] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 10/05/2012] [Accepted: 10/06/2012] [Indexed: 01/08/2023] Open
Abstract
Ginsenoside Rd is a primary constituent of the ginseng rhizome and has been shown to participate in the regulation of diabetes and in tumor formation. Reports also show that ginsenoside Rd exerts anti-oxidative effects by activating anti-oxidant enzymes. Treatment with ginsenoside Rd decreased nitric oxide and prostaglandin E2 (PGE2) in lipopolysaccharides (LPS)-challenged RAW264.7 cells and in ICR mouse livers (5 mg/kg LPS; LPS + ginsenoside Rd [2, 10, and 50 mg/kg]). Furthermore, these decreases were associated with the down-regulations of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 and of nuclear factor (NF)-κB activity in vitro and in vivo. Our results indicate that ginsenoside Rd treatment decreases; 1) nitric oxide production (40% inhibition); 2) PGE2 synthesis (69% to 93% inhibition); 3) NF-κB activity; and 4) the NF-κB-regulated expressions of iNOS and COX-2. Taken together, our results suggest that the anti-inflammatory effects of ginsenoside Rd are due to the down-regulation of NF-κB and the consequent expressional suppressions of iNOS and COX-2.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Kim JS, Kim Y, Han SH, Jeon JY, Hwang M, Im YJ, Kim JH, Lee SY, Chae SW, Kim MG. Development and validation of an LC-MS/MS method for determination of compound K in human plasma and clinical application. J Ginseng Res 2013; 37:135-41. [PMID: 23717167 PMCID: PMC3659617 DOI: 10.5142/jgr.2013.37.135] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/25/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022] Open
Abstract
A rapid, sensitive and selective analytical method was developed and validated for the determination of compound K, a major intestinal bacterial metabolite of ginsenosides in human plasma. Liquid-liquid extraction was used for sample preparation and analysis, followed by liquid chromatography tandem spectrometric analysis and an electrospray-ionization interface. Compound K was analyzed on a Phenomenex Luna C18 column (100×2.00 mm, 3 μm) with the mobile phase run isocratically with 10 mM ammonium acetate-methanol-acetonitrile (5:47.5:47.5, v/v/v) at a flow rate of 0.5 mL/min. The method was validated for accuracy (relative error <12.63%), precision (coefficient of variation <9.14%), linearity, and recovery. The assay was linear over the entire range of calibration standards i.e., a concentration range of 1 ng/mL to 1,000 ng/ mL (r2 >0.9968). The recoveries of compound K after liquid-liquid extraction at 1, 2, 400, and 800 ng/mL were 106.00±0.08%, 103.50±0.19%, 111.45±5.21%, and 89.62±34.46% for intra-day and 85.40±0.08%, 94.50±0.09%, 112.50±5.21%, and 95.87±34.46% for inter-day, respectively. The lower limit of quantification of the analytical method of compound K was 1 ng/ mL in human plasma. The developed method was successfully applied to a pharmacokinetic study of compound K after oral administration in ten of healthy human subjects.
Collapse
Affiliation(s)
- Jung Soo Kim
- Biomedical Research Institute of Chonbuk National University Hospital, Jeonju 561-712, Korea ; Department of Pediatrics, Chonbuk National University Medical School, Jeonju 561-180, Korea ; Research Institute of Clinical Medicine, Chonbuk National University, Jeonju 561-712, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Ginsenoside Rh2 Downregulates LPS-Induced NF- κ B Activation through Inhibition of TAK1 Phosphorylation in RAW 264.7 Murine Macrophage. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:646728. [PMID: 23483870 PMCID: PMC3588194 DOI: 10.1155/2013/646728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 12/27/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022]
Abstract
The present study was carried out to evaluate the inhibitory effects of ginsenoside Rh2 on nuclear-factor- (NF-) κB in lipopolysaccharide- (LPS-) activated RAW 264.7 murine macrophages. RAW 264.7 cells were pretreated with indicated concentrations of ginsenoside Rh2 for 1 h prior to the incubation of LPS (1 μg/mL) for indicated time period. Ginsenoside Rh2 reduced CD14 and Toll-like receptor 4 (TLR4) expressions 24 h after LPS stimulation. Furthermore, ginsenoside Rh2 significantly inhibited TGF-beta-activated kinase 1 (TAK1) phosphorylation 30 min after LPS stimulation. Ginsenoside Rh2 was further shown to inhibit NF-κB p65 translocation into the nucleus by suppressing IκB-α degradation. Also, LPS increased mRNA expression of TNF-α and IL-1α time-dependently, while TQ reduced TNF-α within 3 h and IL-1α within 1 h. And we firstly found that pretreatment of ginsenoside Rh2 successively inhibited hypoxia-inducible factor- (HIF-) 1α expression increased by LPS. In conclusion, ginsenoside Rh2 may inhibit LPS-induced NF-κB activation and reduce HIF-1α accumulation, suggesting that ginsenoside Rh2 may be considered as a potential therapeutic candidate for chronic inflammatory diseases.
Collapse
|
87
|
Park BG, Jung HJ, Cho YW, Lim HW, Lim CJ. Potentiation of antioxidative and anti-inflammatory properties of cultured wild ginseng root extract through probiotic fermentation. ACTA ACUST UNITED AC 2012; 65:457-64. [PMID: 23356855 DOI: 10.1111/jphp.12004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 10/16/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This work aimed to determine some pharmacological properties of non-fermented (WG) and fermented (FWG) extracts of cultured wild ginseng root. METHODS WG was treated with Bifidobacterium longum to generate FWG. Ginsenoside patterns were analysed using thin-layer chromatography and high-performance liquid chromatography. The effect of WG and FWG on reactive oxygen species (ROS) was examined in lipopolysaccharide-stimulated RAW264.7 macrophage cells. Intracellular ROS were detected by flow cytometry. Nitrite in culture supernatant fractions was determined using the Griess reaction. 1,1-Diphenyl-2-picrylhydrazyl was used to determine anti-radical activity. Cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. KEY FINDINGS FWG was rich in ginsenosides Rg3 and Rh2, compared with WG. FWG diminished the enhanced ROS level more strongly than WG in lipopolysaccharide-stimulated RAW264.7 macrophage cells. Both WG and FWG decreased the nitrite levels in stimulated macrophage cells with half-maximal inhibitory concentration (IC50) values of 2.7 and 1.5 mg/ml, respectively, implying that FWG had an enhanced anti-inflammatory activity. Neither WG nor FWG exhibited cytotoxicity on the macrophage cells. In the radical scavenging assay, the IC50 values of WG and FWG were 32.6 and 0.78 mg/ml, respectively, suggesting that FWG had an increased scavenging activity. CONCLUSIONS FWG possesses enhanced antioxidative and anti-inflammatory activity, indicating that fermentation of cultured wild ginseng root extract with a probiotic bacterium can strengthen some of its desirable effects.
Collapse
Affiliation(s)
- Byoung-Gun Park
- Department of Biochemistry, Kangwon National University, Chuncheon, Korea
| | | | | | | | | |
Collapse
|
88
|
Lee JS, Song JH, Sohn NW, Shin JW. Inhibitory Effects of Ginsenoside Rb1 on Neuroinflammation Following Systemic Lipopolysaccharide Treatment in Mice. Phytother Res 2012; 27:1270-6. [DOI: 10.1002/ptr.4852] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/29/2012] [Accepted: 09/02/2012] [Indexed: 12/26/2022]
Affiliation(s)
- Joon-Suk Lee
- Department of Oriental Medical Science, Graduate School of East-west Medical Science; Kyung Hee University; Yongin; 446-701; South Korea
| | | | | | | |
Collapse
|
89
|
Lee IA, Hyam SR, Jang SE, Han MJ, Kim DH. Ginsenoside Re ameliorates inflammation by inhibiting the binding of lipopolysaccharide to TLR4 on macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9595-9602. [PMID: 22849695 DOI: 10.1021/jf301372g] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Ginseng (the root of Panax ginseng C.A. Meyer, family Araliaceae), which contains protopanaxadiol ginsenoside Rb1 and protopanaxatriol ginsenoside Re as main constituents, is frequently used to treat cancer, inflammation, and stress. In the preliminary study, protopanaxatriol ginsenoside Re inhibited NF-κB activation in lipopolysaccharide (LPS)-stimulated murine peritoneal macrophages. Therefore, we investigated its anti-inflammatory effect in peptidoglycan (PGN)-, LPS-, or tumor necrosis factor-α (TNF-α)-stimulated peritoneal macrophages and, in addition, in LPS-induced systemic inflammation and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. Ginsenoside Re inhibited IKK-β phosphorylation and NF-κB activation, as well as the expression of proinflammatory cytokines, TNF-α and IL-1β, in LPS-stimulated peritoneal macrophages, but it did not inhibit them in TNF-α- or PG-stimulated peritoneal macrophages. Ginsenoside Re also inhibited IRAK-1 phosphorylation induced by LPS, as well as IRAK-1 and IRAK-4 degradations in LPS-stimulated peritoneal macrophages. Ginsenoside Re inhibited the binding of Alexa Fluor 488-conjugated LPS to TLR4 on peritoneal macrophages. Furthermore, ginsenoside Re inhibited the binding of LPS to TLR4 on peritoneal macrophages transiently transfected with MyD88 siRNAs. Orally administered ginsenoside Re significantly inhibited the expression of IL-1β and TNF-α on LPS-induced systemic inflammation and TNBS-induced colitis in mice. Ginsenoside Re inhibited colon shortening and myeloperoxidase activity in TNBS-treated mice. Ginsenoside Re reversed the reduced expression of tight-junction-associated proteins ZO-1, claudin-1, and occludin. Ginsenoside Re (20 mg/kg) inhibited the activation of NF-κB in TNBS-treated mice. On the basis of these findings, ginsenoside Re may ameliorate inflammation by inhibiting the binding of LPS to TLR4 on macrophages.
Collapse
Affiliation(s)
- In-Ah Lee
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University , Seoul 130-701, Korea
| | | | | | | | | |
Collapse
|
90
|
|
91
|
Tseng YJ, Shen KP, Lin HL, Huang CY, Dai CF, Sheu JH. Lochmolins A-G, new sesquiterpenoids from the soft coral Sinularia lochmodes. Mar Drugs 2012; 10:1572-1581. [PMID: 22851927 PMCID: PMC3407932 DOI: 10.3390/md10071572] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/06/2012] [Accepted: 07/13/2012] [Indexed: 11/25/2022] Open
Abstract
Seven new sesquiterpenoids, lochmolins A–G (1–7), were isolated from a Taiwanese soft coral Sinularia lochmodes. The structures of these metabolites were elucidated by extensive spectroscopic study. Compounds 1–4 were found to inhibit the accumulation of the LPS-induced pro-inflammatory COX-2 protein in RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Yen-Ju Tseng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (Y.-J.T.); (C.-Y.H.)
| | - Kuo-Ping Shen
- Department of Nursing, Meiho University, Pingtung 912, Taiwan;
| | - Hui-Li Lin
- Department of Food and Nutrition, Meiho University, Pingtung 912, Taiwan;
| | - Chiung-Yao Huang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (Y.-J.T.); (C.-Y.H.)
| | - Chang-Feng Dai
- Institute of Oceanography, National Taiwan University, Taipei 112, Taiwan;
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (Y.-J.T.); (C.-Y.H.)
- Division of Marine Biotechnology, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Author to whom correspondence should be addressed; ; Tel.: +886-7-525-2000 (ext. 5030); Fax: +886-7-525-5020
| |
Collapse
|
92
|
Jang SE, Jung IH, Joh EH, Han MJ, Kim DH. Antibiotics attenuate anti-scratching behavioral effect of ginsenoside Re in mice. JOURNAL OF ETHNOPHARMACOLOGY 2012; 142:105-112. [PMID: 22855946 DOI: 10.1016/j.jep.2012.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Panax ginseng CA Meyer (ginseng) has been used for diabetes, cancer, stress and allergic diseases in the traditional Chinese medicine. AIM OF THE STUDY To understand the role of intestinal microflora in the pharmacological effect of ginsenoside Re, which is a main constituent of ginseng, we investigated its anti-scratching behavioral effect in the mice treated with or without antibiotics. MATERIALS AND METHODS Ginsenoside Re was orally administered to the mice treated with antibiotics (cefadroxil, oxytetracycline and erythromycin mixture (COE), streptomycin or/and tetracycline) and then investigated the relationship between ginsenoside Re-metabolizing β-glucosidase and α-rhamnosidase activities of intestinal microflora and its antiscratching behavioral effect. The anti-scratching behavioral effects of ginsenosides were investigated in the increments of 1 h and 6 h after their oral administrations. The scratching behavioral frequency was measured for 1 h after treatment with histamine. RESULTS Ginsenoside Re inhibited histamine-induced scratching behavior in mice. The anti-scratching behavioral effect of ginsenoside Re was more potent 6 h after its oral administration than 1 h after. However, its inhibitory effect was significantly attenuated in mice treated with COE, but it nearly was not affected in mice treated with streptomycin and/or tetracycline. Treatment with COE also significantly lowered fecal ginsenoside Re-metabolizing β-glucosidase and α-rhamnosidase activities in mice, as well as fecal metabolic activity of ginsenoside Re to ginsenoside Rh1. The anti-scratching behavioral effect of ginsenoside Rh1, a metabolite of ginsenoside Re by intestinal microflora, was superior to that of ginsenoside Re. Ginsenoside Rh1 potently inhibited the expression of IL-4 and TNF-α, as well as the activation of NF-κB and c-jun activation in histamine-stimulated scratching behavioral mice. CONCLUSION Ginsenoside Re may be metabolized to ginsenoside Rh1 by intestinal microflora, which enhances its anti-scratching behavioral effect by inhibiting NF-κB and c-jun activations.
Collapse
Affiliation(s)
- Se-Eun Jang
- Department of Food and Nutrition, Kyung Hee University, 1 Hoegi, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | | | | | | | | |
Collapse
|
93
|
Li R, Yan H, Wang Z, Gong J. Correlation of Solubility and Prediction of the Mixing Properties of Ginsenoside Compound K in Various Solvents. Ind Eng Chem Res 2012. [DOI: 10.1021/ie300945p] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Runyan Li
- School
of Chemical
Engineering and Technology, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Hao Yan
- School
of Chemical
Engineering and Technology, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Zhao Wang
- School
of Chemical
Engineering and Technology, Tianjin University, Tianjin 300072, People’s Republic of China
| | - Junbo Gong
- School
of Chemical
Engineering and Technology, Tianjin University, Tianjin 300072, People’s Republic of China
- Tianjin Key Laboratory
of Modern Drug Delivery and High-Efficiency, Tianjin University, Tianjin 300072, People’s
Republic of China
| |
Collapse
|
94
|
Joh EH, Gu W, Kim DH. Echinocystic acid ameliorates lung inflammation in mice and alveolar macrophages by inhibiting the binding of LPS to TLR4 in NF-κB and MAPK pathways. Biochem Pharmacol 2012; 84:331-40. [PMID: 22564908 DOI: 10.1016/j.bcp.2012.04.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 04/27/2012] [Accepted: 04/27/2012] [Indexed: 12/30/2022]
Abstract
Orally administered lancemaside A, which is isolated from Codonopsis lanceolata (family Campanulaceae), showed anti-colitic effect in mice. However, its metabolite echinocystic acid was absorbed into the blood. Therefore, its anti-inflammatory effects were investigated in lipopolysaccharide (LPS)-stimulated alveolar macrophages in vitro and acute lung injury in vivo. Alveolar macrophages from mice were stimulated with LPS and were treated with echinocystic acid. Acute lung injury was induced by intratracheal administration of LPS in mice. Mice were treated with echinocystic acid or dexamethasone. Echinocystic acid potently suppressed the production of the pro-inflammatory cytokines, TNF-α and IL-1β, as well as of the activations of NF-κB and MAPKS, in LPS-stimulated alveolar macrophages. Echinocystic acid also down-regulated the production of inflammatory markers, which included inducible nitric oxide synthase and cyclooxygenase-2, as well as the inflammatory mediators, nitric oxide and prostaglandin E(2), in LPS-stimulated alveolar macrophages. Echinocystic acid also inhibited the activation of IL-1 receptor-associated kinases, and the activation of mitogen-activated protein kinases in LPS-stimulated alveolar macrophages. Furthermore, echinocystic acid potently inhibited the interaction between LPS and TLR4 in alveolar macrophages transfected with or without MyD88 siRNA, although it did not inhibit the binding in the macrophages transfected with TLR4 siRNA. Echinocystic acid suppressed LPS-induced acute lung inflammation in mice, as well as the expression of pro-inflammatory cytokines, such as IL-1β and TNF-α, and their transcription factor, NF-κB. On the basis of these findings, echinocystic acid, a metabolite of lancemaside A, may express anti-inflammatory effects by inhibiting the binding of LPS to TLR4 on macrophages.
Collapse
Affiliation(s)
- Eun-Ha Joh
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | |
Collapse
|
95
|
Byun MW. Anti-Inflammatory Activity of Austroinulin from Stevia rebaudiana in LPS-induced RAW264.7 Cells. ACTA ACUST UNITED AC 2012. [DOI: 10.3746/jkfn.2012.41.4.456] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
96
|
Jung HJ, Choi H, Lim HW, Shin D, Kim H, Kwon B, Lee JE, Park EH, Lim CJ. Enhancement of anti-inflammatory and antinociceptive actions of red ginseng extract by fermentation. J Pharm Pharmacol 2012; 64:756-62. [DOI: 10.1111/j.2042-7158.2012.01460.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Abstract
Objectives
This work aimed to compare some pharmacological properties of red ginseng extract (RG) and fermented red ginseng extract (FRG).
Methods
Antinociceptive activity was analysed using the acetic acid-induced abdominal constriction response. Anti-inflammatory activity was evaluated using acetic acid-induced vascular permeability and carrageenan-induced inflammation in the air pouch, and analysed through the measurement of nitrite content in the lipopolysaccharide (LPS)-stimulated macrophage cells. Anti-angiogenic activity was determined using the chick chorioallantoic membrane assay.
Key findings
In-vivo anti-inflammatory activity of FRG was stronger than that of RG in two animal models, vascular permeability and air-pouch models. In the vascular permeability model, the doses of RG and FRG required for half-maximal inhibition (IC50) were 181 and 59 mg/kg, respectively. FRG exhibited significantly stronger antinociceptive activity than RG. In the acetic acid-induced abdominal constriction response, the IC50 values of RG and FRG were 153 and 27 mg/kg, respectively. Although both RG and FRG were able to suppress production of nitric oxide in the LPS-stimulated RAW264.7 macrophage cells, the suppressive activity of FRG appeared to be stronger than that of RG. However, RG and FRG showed similar anti-angiogenic activity.
Conclusions
FRG possesses enhanced anti-inflammatory and antinociceptive activity but similar anti-angiogenic activity than RG.
Collapse
Affiliation(s)
- Hyun-Joo Jung
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Hojin Choi
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hye-Won Lim
- Shebah Biotech Inc., Chuncheon Biotechnology Foundation, Hi-Tech Venture Town, Chuncheon, Korea
| | - Daehyun Shin
- R & D Center, Somang Cosmetics Co., Incheon, Korea
| | - Hacksoo Kim
- R & D Team, Seoul Cosmetics Co., Incheon, Korea
| | | | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Eun-Hee Park
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| | - Chang-Jin Lim
- Department of Biochemistry, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
97
|
Wang L, Zhang Y, Wang Z, Li S, Min G, Wang L, Chen J, Cheng J, Wu Y. Inhibitory effect of ginsenoside-Rd on carrageenan-induced inflammation in rats. Can J Physiol Pharmacol 2012; 90:229-36. [PMID: 22300288 DOI: 10.1139/y11-127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A previous study reported that ginsenoside-Rd reduced the production of tumor necrosis factor-α by inhibiting nuclear factor-κB in lipopolysaccharide-activated N9 microglia in vitro. The aim of the present study was to confirm the anti-inflammatory effects and mechanisms of ginsenoside-Rd in animal experiments involving acute inflammation. The results indicated that ginsenoside-Rd at doses ranging from 12.5 to 50 mg/kg i.m. significantly inhibited the swelling of hind paws in rats for 1-6 h after the carrageenan injection. The levels of proinflammatory cytokines and proinflammatory mediators were markedly reduced by ginsenoside-Rd. Ginsenoside-Rd, when administered intramuscularly at 12.5, 25, and 50 mg/kg doses, showed signicant inhibition of carrageenan-induced production of interleukin-1β (6.91%, 45.75%, and 55.18%, respectively), tumor necrosis factor-α (37.99%, 56.39%, and 47.38%, respectively), prostaglandin E(2) (22.92%, 30.12%, and 36.36%, respectively), and nitric oxide (28.27%, 44.53%, and 53.42%, respectively). In addition, ginsenoside-Rd (12.5, 25, and 50 mg/kg i.m.) effectively decreased the levels of nuclear factor-κB (6.77%, 20.28%, and 41.03%, respectively) and phosphorylation of IκBα (13.23%, 26.92%, and 41.80%, respectively) in the carrageenan-inflamed paw tissues. These results suggest that ginsenoside-Rd has significant anti-inflammatory effects in vivo, which might be due to its blocking of the nuclear factor-κB signaling pathway. Thus, it may be possible to develop ginsenoside-Rd as a useful agent for inflammatory diseases.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Li R, Dong W, He H, Yan H, Jiang X, Gong J. Isolation, characterization and phase transformation of new ginsenoside compound k hydrate and methanol solvates. CRYSTAL RESEARCH AND TECHNOLOGY 2012. [DOI: 10.1002/crat.201200016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
99
|
Shih HJ, Tseng YJ, Huang CY, Wen ZH, Dai CF, Sheu JH. Cytotoxic and anti-inflammatory diterpenoids from the Dongsha Atoll soft coral Sinularia flexibilis. Tetrahedron 2012. [DOI: 10.1016/j.tet.2011.10.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
100
|
Jang YJ, Kim ME, Ko SY. n-Butanol extracts of Panax notoginseng suppress LPS-induced MMP-2 expression in periodontal ligament fibroblasts and inhibit osteoclastogenesis by suppressing MAPK in LPS-activated RAW264.7 cells. Arch Oral Biol 2011; 56:1319-27. [DOI: 10.1016/j.archoralbio.2011.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 05/19/2011] [Accepted: 05/23/2011] [Indexed: 11/29/2022]
|