Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
2.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119461, 2023 06.
Article in English | MEDLINE | ID: mdl-36931607

ABSTRACT

As an anticancer drug, cisplatin is widely used, but its clinical application is restricted due to its severe side effects of ototoxicity. Therefore, this study was dedicated to assessing the benefit of ginsenoside extract, 20(S)-Ginsenoside Rh1 (Rh1), on cisplatin-induced ototoxicity. HEI-OC1 cells and neonatal cochlear explants were cultured. Cleaved caspase-3, TUNEL, and MitoSOX Red were observed in vitro by immunofluorescence staining. CCK8 and LDH cytotoxicity assays were detected to measure cell viability and cytotoxicity. Our results showed that Rh1 significantly increased cell viability, reduced cytotoxicity, and alleviated cisplatin-induced apoptosis. In addition, Rh1 pretreatment decreased the excessive accumulation of intracellular reactive oxygen species. Mechanistic studies indicated that Rh1 pretreatment reversed the increase of apoptotic protein expression, accumulation of mitochondrial ROS, and activation of the MAPK signaling pathway. These results suggested that Rh1 can act as an antioxidant and anti-apoptotic agent against cisplatin-induced hearing loss by suppressing the excessive accumulation of mitochondrial ROS, activation of MAPK signaling pathway and apoptosis.


Subject(s)
Ginsenosides , Hearing Loss , Ototoxicity , Humans , Infant, Newborn , Cisplatin/adverse effects , Ginsenosides/adverse effects , Ginsenosides/metabolism , Reactive Oxygen Species/metabolism , Ototoxicity/metabolism , Hair Cells, Auditory , Hearing Loss/chemically induced , Apoptosis , MAP Kinase Signaling System
3.
Environ Sci Pollut Res Int ; 30(12): 34188-34202, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36508105

ABSTRACT

Tibial dyschondroplasia (TD) is a developmental cartilaginous disease due to thiram toxicity. The abnormity of chondrocytes and insufficient angiogenesis within the growth plate are the major factors leading to the occurrence of TD in most cases. In the current study, we evaluated the beneficial effects of ginsenoside (Rg1) against thiram-induced TD for knowing the possible underlying mechanisms in broiler chickens through in vivo and in vitro assessment. Arbor acres broilers (1-day-old, n = 120) were randomly divided for the in vivo evaluation. The control broilers were fed under normal conditions during the whole experiment cycle (18 days). The TD broilers were fed with 50 mg/kg thiram, while the treatment group was given 40 mg/kg of Rg1. According to our findings, thiram caused a decrease in production performance and tibia parameters (p < 0.05), which were significantly reversed by Rg1 administration. In addition, the results from the histological evaluation showed that the proliferative zone had a smaller number of blood vessels, surrounded by inviable chondrocytes, proving apoptosis during the occurrence of TD, while Rg1 treatment significantly increased blood vessels and decreased apoptotic cells. Furthermore, it was found that Rg1 effectively ameliorated the angiogenesis by regulation of HIF-1α/VEGFA/VEGFR2 signaling pathway and the chondrocytes' apoptosis via the mitochondrial pathway. Hence, these findings suggest that Rg1 might be a perfect choice in the prevention and treatment of TD via regulating chondrocytes apoptosis and angiogenesis. Also, it might be a potential therapeutic drug for humans to overcome different bone disorders, involving chondrocytes.


Subject(s)
Ginsenosides , Osteochondrodysplasias , Humans , Animals , Thiram/toxicity , Chickens , Ginsenosides/adverse effects , Chondrocytes/pathology , Apoptosis , Osteochondrodysplasias/veterinary
4.
Int J Med Sci ; 19(1): 65-73, 2022.
Article in English | MEDLINE | ID: mdl-34975299

ABSTRACT

Reperfusion injury following myocardial ischemia remained a challenge for optimal treatment of myocardial infarction. Ginsenosides Rb (G-Rb), the primary components of ginsenoside, have been reported to exert cardioprotective effects via numerous mechanisms. G-Rb1 mediate cardioprotective effects via various signaling pathways, including mitochondrial apoptotic pathway, PI3K/Akt/mTOR, HIF-1α and GRF91, RhoA, p38α MAPK, and eNOS. G-Rb2 activates the SIRT-1 pathway, while G-Rb3 promotes both JNK-mediated NF-κB and PERK/Nrf2/HMOX1. Generally, ginsenosides Rb1, 2, and 3 modulates oxidative stress, inflammation, and apoptosis, contributing to the improvement of structural, functional and biochemical parameters. In conclusion, G-Rb, particularly G-Rb1, have vast potential as a supplement in attenuating reperfusion injury. Translation into a clinical trial is warranted to confirm the beneficial effects of G-Rb.


Subject(s)
Ginsenosides/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis , Cardiotonic Agents/adverse effects , Cardiotonic Agents/therapeutic use , Ginsenosides/adverse effects , Ginsenosides/therapeutic use , Inflammation/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress , Signal Transduction
5.
Immunopharmacol Immunotoxicol ; 44(1): 110-118, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34898349

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is dramatically increasing worldwide, cannot be thoroughly cured, and reduces patients' quality of life. Excessive activation of macrophages and over-production of cytokines play an important role in the pathogenesis of UC. Therefore, for its treatment, inhibiting macrophages' hyperactivation would be effective to develop new treatment approaches. Ginsenosides, extracted from ginseng, show an anti-inflammatory effect on the immunologic process. Our study used ginsenosides Rd monomer (GRd) to intervene in DSS-induced colitis mouse models and tested the immunological effect of macrophages. METHOD: We observed body weights, weights of colons, colonic lengths, and inflammatory scores, as well as histological changes of DSS/DSS-GRd mice. We also isolated intestinal and peritoneal macrophages, performed qRT-PCR and ELISA to detect cytokines production by macrophages, and screened possible involved pathways by Western blotting. RESULTS: Administering 20 mg/Kg GRd to DSS mice for 7-14 days reduced colonic inflammation. Moreover, both in vivo and in vitro, levels of TNF-α, IFN-γ, IL-6, IL-12/23p40, and IL-17A were all inhibited by GRd at 14 days in intestinal macrophages, and 20 µmol/L GRd at 12 h in peritoneal macrophages, respectively, but longer time made no more benefit. Western blotting showed GRd could decrease expression of pJNK, p-p38, pIκBα, and P65 in nuclear. CONCLUSIONS: Our data indicate that GRd could down-regulate cytokines production in macrophages and alleviate DSS-colitis in mice, which may be related to NF-κB and P38MAPK pathways. These results suggest that GRd has an anti-inflammatory effect on experimental colitis and may have potential efficacy in the treatment of UC alone or in combination.


Subject(s)
Colitis, Ulcerative , Colitis , Ginsenosides , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis, Ulcerative/chemically induced , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Ginsenosides/adverse effects , Ginsenosides/metabolism , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Quality of Life , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Pharmacol Res ; 166: 105481, 2021 04.
Article in English | MEDLINE | ID: mdl-33549726

ABSTRACT

Cardiovascular disease (CVD) remains the major cause of death worldwide, accounting for almost 31% of the global mortality annually. Several preclinical studies have indicated that ginseng and the major bioactive ingredient (ginsenosides) can modulate several CVDs through diverse mechanisms. However, there is paucity in the translation of such experiments into clinical arena for cardiovascular ailments due to lack of conclusive specific pathways through which these activities are initiated and lack of larger, long-term well-structured clinical trials. Therefore, this review elaborates on current pharmacological effects of ginseng and ginsenosides in the cardiovascular system and provides some insights into the safety, toxicity, and synergistic effects in human trials. The review concludes that before ginseng, ginsenosides and their preparations could be utilized in the clinical treatment of CVDs, there should be more preclinical studies in larger animals (like the guinea pig, rabbit, dog, and monkey) to find the specific dosages, address the toxicity, safety and synergistic effects with other conventional drugs. This could lead to the initiation of large-scale, long-term well-structured randomized, and placebo-controlled clinical trials to test whether treatment is effective for a longer period and test the efficacy against other conventional therapies.


Subject(s)
Cardiotonic Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Ginsenosides/therapeutic use , Animals , Cardiotonic Agents/adverse effects , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/pathology , Ginsenosides/adverse effects , Ginsenosides/chemistry , Ginsenosides/pharmacology , Humans , Panax/chemistry , Phytotherapy
7.
Pharmacol Res ; 159: 105031, 2020 09.
Article in English | MEDLINE | ID: mdl-32562816

ABSTRACT

Thrombosis initiated by abnormal platelet aggregation is a pivotal pathological event that precedes most cases of cardiovascular diseases (CVD). Recently, growing evidence indicates that platelet could be a potential target for CVD prevention. However, as the conventional antithrombotic management strategy, applications of current antiplatelet agents are somewhat limited by their various side effects, such as bleeding risk and drug resistance. Hence, efforts have been made to search for agents as complementary therapies. Ginsenoside, the principal active component extracted from Panax ginseng, has gained much attention for its regulations on multiple crucial events of platelet aggregation. From structural characteristics to clinical applications, this review anatomized the intrinsic structure-function relationship of antiplatelet potency of ginsenosides, and the involved signal pathways were specifically summarized. Additionally, the emphasis was placed on clinical studies that investigate the antithrombotic efficacy of ginsenosides in the treatment of CVD. Further, a broad overview of approaches for improving the bioavailability of ginsenosides was concluded. Limitations and prospects of current studies were also discussed. This study may provide some new insights into the systematic understanding of ginsenosides in CVD treatment and lay a foundation for future research.


Subject(s)
Blood Platelets/drug effects , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Ginsenosides/therapeutic use , Muscle, Smooth, Vascular/drug effects , Neointima , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Vascular Remodeling/drug effects , Animals , Biological Availability , Blood Platelets/metabolism , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Ginsenosides/adverse effects , Ginsenosides/pharmacokinetics , Humans , Molecular Structure , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Signal Transduction , Structure-Activity Relationship
8.
Phytother Res ; 34(7): 1659-1669, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32100342

ABSTRACT

Clinical dose of doxorubicin (100 nM) induced cellular senescence and various secretory phenotypes in breast cancer and normal epithelial cells. Herein, we reported the detailed mechanism underlying ginsenoside Rh2-mediated NF-κB inhibition, and mitophagy promotion were evaluated by antibody array assay, western blotting analysis, and immunocytostaining. Ginsenoside Rh2 suppressed the protein levels of TRAF6, p62, phosphorylated IKK, and IκB, which consequently inactivated NF-κB activity. Rh2-mediated secretory phenotype was delineated by the suppressed IL-8 secretion. Senescent epithelial cells showed increased level of reactive oxygen species (ROS), which was significantly abrogated by Rh2, with upregulation on SIRT 3 and SIRT 5 and subsequent increase in SOD1 and SOD2. Rh2 remarkably favored mitophagy by the increased expressions of PINK1 and Parkin and decreased level of PGC-1α. A decreased secretion of IL-8 challenged by mitophagy inhibitor Mdivi-1 with an NF-κB luciferase system was confirmed. Importantly, secretory senescent epithelial cells promoted the breast cancer (MCF-7) proliferation while decreased the survival of normal epithelial cells demonstrated by co-culture system, which was remarkably alleviated by ginsenoside Rh2 treatment. These data included ginsenoside Rh2 regulated ROS and mitochondrial autophagy, which were in large part attributed to secretory phenotype of senescent breast epithelial cells induced by doxorubicin. These findings also suggested that ginsenoside Rh2 is a potential treatment candidate for the attenuation of aging related disease.


Subject(s)
Breast Neoplasms/chemically induced , Doxorubicin/adverse effects , Drugs, Chinese Herbal/therapeutic use , Ginsenosides/adverse effects , Mitochondria/metabolism , Autophagy , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Line, Tumor , Drugs, Chinese Herbal/pharmacology , Female , Humans , Oxidative Stress
9.
Molecules ; 24(18)2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31505740

ABSTRACT

Pien-Tze-Huang (PTH) is a famous and commonly used traditional Chinese medicine formula in China. It was first formulated by a royal physician of the Ming Dynasty (around 1555 AD). Recently, PTH has attracted attention worldwide due to its beneficial effects against various diseases, especially cancer. This paper systematically reviewed the up-to-date information on its chemical composition, pharmacology, and clinical application. A range of chemical compounds, mainly ginsenosides and bile acids, have been identified and quantified from PTH. Pharmacological studies indicated that PTH has beneficial effects against various cancers, hepatopathy, and ischemic stroke. Furthermore, PTH has been used clinically to treat various diseases in China, such as colorectal cancer, liver cancer, and hepatitis. In summary, PTH is a potential agent with extensive therapeutic effects for the treatment of various diseases. However, the lack of information on the side effects and toxicity of PTH is a non-negligible issue, which needs to be seriously studied in the future.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Neoplasms/drug therapy , Stroke/drug therapy , Bile Acids and Salts/adverse effects , Bile Acids and Salts/therapeutic use , Drugs, Chinese Herbal/adverse effects , Ginsenosides/adverse effects , Ginsenosides/therapeutic use , Humans , Medicine, Chinese Traditional/adverse effects
10.
Food Chem Toxicol ; 131: 110553, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31163221

ABSTRACT

Ginseng and its active gradient, ginsenoside Rg3 (Rg3), are widely used for a variety of health benefits, but concerns over their misuses are increasing. Previously, it has been reported that Rg3 can cause hemolysis, but its health outcome remains unknown. Here, we demonstrated that Rg3 could promote the procoagulant activity of erythrocytes through the process of hemolysis, ultimately leading to increased thrombosis. In freshly isolated human erythrocytes, Rg3 caused pore formation and fragmentation of the erythrocyte membrane. Confocal microscopy observation and flow cytometric analysis revealed that remnant erythrocyte fragments after the exposure to Rg3 expressed phosphatidylserine (PS), which can promote blood coagulation through providing assembly sites for coagulation complexes. Rat in vivo experiments further confirmed that intravenous administration of Rg3 produced PS-bearing erythrocyte debris and increased thrombosis. Collectively, we demonstrated that Rg3 could induce the procoagulant activity of erythrocytes by generating PS-bearing erythrocyte debris through hemolysis, which might provoke thrombosis.


Subject(s)
Blood Coagulation/drug effects , Erythrocytes/drug effects , Ginsenosides/adverse effects , Hemolysis/drug effects , Thrombosis/chemically induced , Animals , Erythrocyte Membrane/chemistry , Erythrocyte Membrane/drug effects , Erythrocytes/metabolism , Erythrocytes/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Phosphatidylserines/chemistry , Rats, Sprague-Dawley
11.
Regul Toxicol Pharmacol ; 92: 83-93, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29155077

ABSTRACT

Ginseng is a major herbal remedy used in Asian countries for thousands of years and known to restore and enhance vital energy. Korean red ginseng, which is processed by steaming and drying fresh Panax ginseng, is most popular and contains unique ginsenosides, which have anticancer and anti-inflammatory properties. The present study was carried out to evaluate the repeated oral dose toxicity of Korean red ginseng extract. The test article was administered orally once a day to male and female Sprague-Dawley rats at dose levels of 0, 500, 1000, or 2000 mg/kg/day for 13 consecutive weeks (15 animals/sex/group in the vehicle control and 2000 mg/kg/day groups, and 10 animals/sex/group in the 500 and 1000 mg/kg/day groups). Ten animals per group were sacrificed at the end of the 13-week treatment period, and the remaining rats were sacrificed after a 4-week recovery period. Administration of Korean red ginseng extract did not result in any toxicologically significant changes in mortality, body weight, food consumption, ophthalmoscopy, hematology, serum biochemistry, gross pathological findings, absolute/relative organ weights, or histopathology. It was established that the no observed adverse effect level (NOAEL) of the test article was 2000 mg/kg/day for both sexes in this study.


Subject(s)
Panax/adverse effects , Plant Extracts/adverse effects , Administration, Oral , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Ginsenosides/adverse effects , Korea , Male , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Rats , Rats, Sprague-Dawley
12.
Fitoterapia ; 121: 223-228, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28734737

ABSTRACT

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


Subject(s)
Ginsenosides/adverse effects , Hypersensitivity/physiopathology , Mast Cells/drug effects , Animals , Cell Line, Tumor , Histamine/metabolism , Humans , Hypersensitivity/metabolism , Male , Mast Cells/metabolism , Mice , Mice, Inbred ICR , Molecular Structure , Phosphatidylserines/metabolism , Rats , beta-N-Acetylhexosaminidases/metabolism
13.
Can J Physiol Pharmacol ; 95(10): 1170-1176, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28505464

ABSTRACT

Heart failure is a major medical and economic burden throughout the world. Although various treatment options are available to treat heart failure, death rates in both men and women remain high. Potential adjunctive therapies may lie with use of herbal medications, many of which possess potent pharmacological properties. Among the most widely studied is ginseng, a member of the genus Panax that is grown in many parts of the world and that has been used as a medical treatment for a variety of conditions for thousands of years, particularly in Asian societies. There are a number of ginseng species, each possessing distinct pharmacological effects due primarily to differences in their bioactive components including saponin ginsenosides and polysaccharides. While experimental evidence for salutary effects of ginseng on heart failure is robust, clinical evidence is less so, primarily due to a paucity of large-scale well-controlled clinical trials. However, there is evidence from small trials that ginseng-containing Chinese medications such as Shenmai can offer benefit when administered as adjunctive therapy to heart failure patients. Substantial additional studies are required, particularly in the clinical arena, to provide evidence for a favourable effect of ginseng in heart failure patients.


Subject(s)
Cardiomegaly/drug therapy , Cardiovascular Agents/therapeutic use , Ginsenosides/therapeutic use , Heart Failure/drug therapy , Panax/chemistry , Plant Extracts/therapeutic use , Animals , Cardiomegaly/diagnosis , Cardiomegaly/physiopathology , Cardiovascular Agents/adverse effects , Cardiovascular Agents/isolation & purification , Cells, Cultured , Clinical Trials as Topic , Disease Models, Animal , Ginsenosides/adverse effects , Ginsenosides/isolation & purification , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Phytotherapy , Plant Extracts/adverse effects , Plant Extracts/isolation & purification , Plants, Medicinal , Treatment Outcome
14.
Oncotarget ; 7(43): 70535-70545, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27655708

ABSTRACT

PURPOSE: Acquired resistance is a bottleneck that restricts the efficacy of epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) for lung cancer. Ginsenoside Rg3 is an antiangiogenic agent which can down-regulate the expressions of vascular endothelial growth factor (VEGF) and EGFR. Combination of EGFR-TKI and ginsenoside Rg3 may be a promising strategy to delay acquired resistance. This retrospective study explored the efficacy and safety of this combined regimen in patients with EGFR mutation and advanced non-small cell lung cancer (NSCLC). RESULTS: By the deadline of March 31th 2016, the median follow-up period reached 22.9 months. The median PFS was significantly longer in group A than in group B (12.4 months vs 9.9 months, P = 0.017). In addition, ORR was significantly higher in group A than in group B (59.6% vs 41.7%, P = 0.049). The median OS in group A showed no extended tendency compared with that in group B (25.4 months vs 21.4 months, P = 0.258). No significant difference in side effects was found between the two groups. METHODS: A total of 124 patients with advanced NSCLC and EGFR active mutation were collected and analyzed. All of them were treated with first-line EGFR-TKI and divided into two groups. In group A (n=52), patients were administered EGFR-TKI plus ginsenoside Rg3 at standard doses. In group B (n=72), patients received EGFR-TKI alone. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and side effects were analyzed. CONCLUSIONS: Ginsenoside Rg3 improves median PFS and ORR of first-line EGFR-TKI treatment in EGFR-mutant advanced NSCLC patients, thus providing a new regimen to delay acquired resistance of EGFR-TKI.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Mutation , Adult , Aged , Aged, 80 and over , Anorexia/chemically induced , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Exanthema/chemically induced , Female , Ginsenosides/administration & dosage , Ginsenosides/adverse effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Vascular Endothelial Growth Factor A/genetics
15.
Mol Neurobiol ; 53(4): 2529-40, 2016 May.
Article in English | MEDLINE | ID: mdl-26081140

ABSTRACT

A great deal of attention has been paid to neuroprotective therapies for cerebral ischemic stroke. Our two recent clinical trials showed that ginsenoside Rd (Rd), a kind of monomeric compound extracted from Chinese herbs, Panax ginseng and Panax notoginseng, was safe and efficacious for the treatment of ischemic stroke. In this study, we conducted a pooled analysis of the data from 199 patients with acute ischemic stroke in the first trial and 390 in the second to reanalyze the efficacy and safety of Rd. Moreover, animal stroke models were carried out to explore the possible molecular mechanisms underlying Rd neuroprotection. The pooled analysis showed that compared with placebo group, Rd could improve patients' disability as assessed by modified Rankin Scale (mRS) score on day 90 post-stroke and reduce neurologic deficits on day 15 or day 90 post-stroke as assessed by NIH Stroke Scale (NIHSS) and Barthel Index (BI) scores. For neuroprotective mechanisms, administration of Rd 4 h after stroke could inhibit ischemia-induced microglial activation, decrease the expression levels of various proinflammatory cytokines, and suppress nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha (IκBα) phosphorylation and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear translocation. An in vitro proteasome activity assay revealed a significant inhibitory effect of Rd on proteasome activity in microglia. Interestingly, Rd was showed to have less side effects than glucocorticoid. Therefore, our study demonstrated that Rd could safely improve the outcome of patients with ischemic stroke, and this therapeutic effect may result from its capability of suppressing microglial proteasome activity and sequential inflammation.


Subject(s)
Brain Ischemia/drug therapy , Ginsenosides/therapeutic use , Inflammation/pathology , Microglia/pathology , Proteasome Endopeptidase Complex/metabolism , Stroke/drug therapy , Stroke/pathology , Animals , Animals, Newborn , Brain Ischemia/complications , Brain Ischemia/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/metabolism , Dexamethasone/pharmacology , Ginsenosides/adverse effects , Ginsenosides/pharmacology , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Rats, Sprague-Dawley , Stroke/complications , Treatment Outcome
16.
Eur J Drug Metab Pharmacokinet ; 41(6): 845-853, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26470874

ABSTRACT

BACKGROUND AND OBJECTIVES: 20(S)-Ginsenoside Rg3 could significantly inhibit tumor growth and metastasis in animals and in in vitro tumor cell invasion. This first-in-human pharmacokinetic study investigated the pharmacokinetics of 20(S)-ginsenoside Rg3 (hip intramuscular injection) in healthy Chinese volunteers. METHODS: Study 1 investigated single, ascending intramuscular doses of 10-60 mg of 20(S)-ginsenoside Rg3 in 24 healthy adults; study 2 evaluated multiple intramuscular doses of 30 mg of 20(S)-ginsenoside Rg3 administered for 15 days in 9 healthy adults. RESULTS: In both studies, 20(S)-ginsenoside Rg3 was rapidly absorbed, with a time to reach maximum plasma concentration (T max) of 4 h. After single-dose administration, elimination half-life t ½ was 32.0 ± 26.7, 51.7 ± 15.4 and 53.9 ± 25.7 h; maximum plasma concentration (C max) was 135.4 ± 35.3, 162.1 ± 47.2 and 399.8 ± 217.0 ng/mL; area under the plasma concentration-time curve (AUC) from time zero to infinity (AUC0-∞) was 3474.1 ± 1312.3, 8156.5 ± 1782.7 and 25,666.8 ± 9401.1 ng·h/mL; clearance CL/F was 3.2 ± 0.9, 3.8 ± 0.7 and 2.7 ± 1.3 L/h; urine excretion percentage during 72 h was 0.5 ± 0.1, 0.4 ± 0.1 and 0.4 ± 0.2 % after 10, 30 and 60 mg 20(S)-ginsenoside Rg3, respectively. After multiple-dose administration, C max was 457.0 ± 165.7 and 770.2 ± 275.4 ng/mL; AUC0-48h was 10,530.0 ± 4073.9 and 16,871.3 ± 6939.3 ng·h/mL after the first and the last 20(S)-ginsenoside Rg3 doses, respectively; fluctuation percentage was 183.0 ± 46.3 %. Accumulation ratio was 1.7 ± 0.6 at steady state. CONCLUSIONS: 20(S)-ginsenoside Rg3 was generally well tolerated. In these studies, 20(S)-ginsenoside Rg3 exhibited a pharmacokinetic profile suitable for once-every-2-days dosing.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Drugs, Chinese Herbal/pharmacokinetics , Ginsenosides/pharmacokinetics , Immunologic Factors/pharmacokinetics , Absorption, Physiological , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/metabolism , Area Under Curve , Asian People , Buttocks , China , Cohort Studies , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/metabolism , Female , Ginsenosides/administration & dosage , Ginsenosides/adverse effects , Ginsenosides/metabolism , Half-Life , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/metabolism , Injections, Intramuscular , Male , Metabolic Clearance Rate , Renal Elimination , Reproducibility of Results , Urine/chemistry
17.
Arch Pharm Res ; 37(9): 1183-92, 2014.
Article in English | MEDLINE | ID: mdl-24687256

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.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anticarcinogenic Agents/pharmacology , Endothelium, Vascular/drug effects , Ginsenosides/pharmacology , Neovascularization, Pathologic/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Angiogenesis Inhibitors/adverse effects , Anticarcinogenic Agents/adverse effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Ceramides/agonists , Ceramides/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Ginsenosides/adverse effects , Ginsenosides/pharmacokinetics , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Osmolar Concentration , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/agonists , Sphingosine/analogs & derivatives , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Sphingosine/pharmacology
18.
Toxicol Appl Pharmacol ; 275(2): 163-75, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24380838

ABSTRACT

Modulation of drug transporters via herbal medicines which have been widely used in combination with conventional prescription drugs may result in herb-drug interactions in clinical practice. The present study was designed to investigate the inhibitory effects of 50 major herbal constituents on P-glycoprotein (P-gp) in vitro and in vivo as well as related inhibitory mechanisms. Among these herbal medicines, four constituents, including emodin, 18ß-glycyrrhetic acid (18ß-GA), dehydroandrographolide (DAG), and 20(S)-ginsenoside F1 [20(S)-GF1] exhibited significant inhibition (>50%) on P-gp in MDR1-MDCKII and Caco-2 cells. Emodin was the strongest inhibitor of P-gp (IC50=9.42 µM), followed by 18ß-GA (IC50=21.78 µM), 20(S)-GF1 (IC50=76.08 µM) and DAG (IC50=77.80 µM). P-gp ATPase activity, which was used to evaluate the affinity of substrates to P-gp, was stimulated by emodin and DAG with Km and Vmax values of 48.61, 29.09 µM and 71.29, 38.45 nmol/min/mg protein, respectively. However, 18ß-GA and 20(S)-GF1 exhibited significant inhibition on both basal and verapamil-stimulated P-gp ATPase activities at high concentration. Molecular docking analysis (CDOCKER) further elucidated the mechanism for structure-inhibition relationships of herbal constituents with P-gp. When digoxin was co-administered to male SD rats with emodin or 18ß-GA, the AUC(0₋t) and Cmax of digoxin were increased by approximately 51% and 58%, respectively. Furthermore, 18ß-GA, DAG, 20(S)-GF1 and Rh1 at 10 µM significantly inhibited CYP3A4/5 activity, while emodin activated the metabolism of midazolam in human liver microsomes. In conclusion, four herbal constituents demonstrated inhibition of P-gp to specific extents in vitro and in vivo. Taken together, our findings provided the basis for the reliable assessment of the potential risks of herb-drug interactions in humans.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Diterpenes/adverse effects , Emodin/adverse effects , Ginsenosides/adverse effects , Glycyrrhetinic Acid/analogs & derivatives , Herb-Drug Interactions , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Animals , Area Under Curve , Caco-2 Cells , Cell Line, Tumor , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Digoxin/administration & dosage , Glycyrrhetinic Acid/adverse effects , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/pathology , Protein Conformation , Rats , Verapamil/administration & dosage
19.
Planta Med ; 79(12): 1043-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23839821

ABSTRACT

Xue-Sai-Tong injection, a traditional Chinese medicine with total saponins of Sanqi ginseng as active ingredients, has been used for more than 500 years to treat coronary artery disease in China. Anaphylactoid reaction induced by Xue-Sai-Tong injection was one of the main adverse drug reactions which has occurred frequently in recent years. It is of importance to elucidate its anaphylactoid constituents. The in vivo anaphyalctoid tests indicated that the anaphylactoid mediators could be used as indexes to evaluate the anaphylactoid action. Further, the in vitro model based on determining the mediators release from the degranulation of mast cells and RBL-2H3 cells stimulated by Xue-Sai-Tong injection was explored. Mediators released from mast cells and RBL-2H3 cells caused by Xue-Sai-Tong injection were determined by comparison of the methods of fluorospectrophotometry, ELISA, and spectrophotometry, respectively, revealing that the histamine release induced by the Xue-Sai-Tong injection could not be assayed accurately by the method of fluorospectrophotometry because of the interference of saponins and unknown components in the injection. The rat peritoneal mast cell was also not an optimal cell model for determining histamine and ß-hexosaminidase release due to the higher spontaneous release ratio during the cell collection. Thus, ELISA determination of the histamine release from RBL-2H3 cells is a suitable in vitro model to assay the anaphylactoid reaction of Xue-Sai-Tong injection. Previously, abnormal hemolysis in some batches of Xue-Sai-Tong injection was observed in the course of their HD50 (half hemolytic dosage) determination. This study further found that injections which exhibited an abnormal hemolysis phenomenon also caused a higher release of the anaphylactoid mediators from RBL-2H3 cells, indicating the HD50 could be an auxiliary index to evaluate anaphylactoid action of the herbal injection indirectly. Research for anaphylactoid components in Xue-Sai-Tong injection indicated that proteins with over 10 KDa of molecular weight, but not ginsenosides, could be the main constituents inducing the release of anaphylactoid mediators from RBL-2H3 cells. An HPLC method for protein determination in the Xue-Sai-Tong injection was established subsequently, and the content of proteins with molecular weights of over 10 KDa in the injections showed an obviously positive correlation with the histamine release induced by the injections. In addition, taking ginsenoside-Rd coupled with BSA as an example, the hapten property of ginsenosides was studied and the ratio of ginsenoside-Rd to BSA was determined to be 8:1 by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, and the ginsenoside-BSA conjugate showed a stronger action to stimulate histamine release from the RBL-2H3 cells.


Subject(s)
Anaphylaxis/chemically induced , Drugs, Chinese Herbal/adverse effects , Ginsenosides/adverse effects , Panax notoginseng/chemistry , Saponins/adverse effects , Animals , Cell Line, Tumor , China , Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/isolation & purification , Ginsenosides/chemistry , Ginsenosides/isolation & purification , Guinea Pigs , Histamine/metabolism , Histamine Release/drug effects , Male , Mast Cells/drug effects , Mast Cells/metabolism , Medicine, Chinese Traditional , Plant Proteins/analysis , Plant Proteins/metabolism , Rats , Rats, Wistar , Saponins/chemistry , Saponins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , beta-N-Acetylhexosaminidases/drug effects , beta-N-Acetylhexosaminidases/metabolism
20.
Environ Toxicol Pharmacol ; 31(3): 397-405, 2011 May.
Article in English | MEDLINE | ID: mdl-21787710

ABSTRACT

Antitumor effects of a ginsenoside Rg(3)-fortified red ginseng preparation (Rg(3)-RGP) were investigated in human non-small cell lung carcinoma (H460) cells using in vitro cytotoxicity assay and in vivo nude mouse xenograft model. Immunomodulatory effects of the preparation were also assessed by measuring the facilitating activities on the nitric oxide (NO) release from peritoneal macrophages, in vitro and in vivo lymphocyte proliferation, and the carbon clearance from circulating blood. In a cell level, Rg(3)-RGP exerted H460 cytotoxicity and facilitated splenocyte proliferation at very high concentrations, without affecting NO production. However, oral administration of Rg(3)-RGP (100-300 mg/kg) enhanced carbon particle-phagocytic index of blood macrophages up to 360-397% of control value. In addition, Rg(3)-RGP significantly increased the splenocyte proliferation (23% at 100mg/kg). In tumor-bearing mice, 28-day oral treatment with Rg(3)-RGP (100mg/kg) remarkably suppressed the tumor growth, leading to the decrease of the tumor volume and weight by 30-31%, which was comparable to the effect (27-29% reduction) of doxorubicin (2mg/kg at 3-day intervals). While Rg(3)-RGP did not cause adverse effects, intravenous injection of doxorubicin markedly decreased body and testes weights, and exhibited severe depletion of spermatogenic cells in the atrophic seminiferous tubules. These results indicate that Rg(3)-RGP exerts antitumor activities via indirect immunomodulatory actions, without causing adverse effects as seen in doxorubicin.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Ginsenosides/pharmacology , Panax/chemistry , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/adverse effects , Body Weight/drug effects , Carbon/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Ginsenosides/adverse effects , Heart/drug effects , Humans , Lung Neoplasms/drug therapy , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Neoplasm Transplantation , Nitric Oxide/biosynthesis , Organ Size/drug effects , Plant Preparations , Spleen/cytology , Spleen/drug effects , Testis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL