Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Front Nutr ; 10: 1136458, 2023.
Article in English | MEDLINE | ID: mdl-37006921

ABSTRACT

Selenium is an essential microelement involved in various biological processes. Selenium deficiency increases the risk of human immunodeficiency virus infection, cancer, cardiovascular disease, and inflammatory bowel disease. Selenium possesses anti-oxidant, anti-cancer, immunomodulatory, hypoglycemic, and intestinal microbiota-regulating properties. The non-linear dose-response relationship between selenium status and health effects is U-shaped; individuals with low baseline selenium levels may benefit from supplementation, whereas those with acceptable or high selenium levels may face possible health hazards. Selenium supplementation is beneficial in various populations and conditions; however, given its small safety window, the safety of selenium supplementation is still a subject of debate. This review summarizes the current understanding of the health-promoting effects of selenium on the human body, the dietary reference intake, and evidence of the association between selenium deficiency and disease.

2.
Phytomedicine ; 110: 154652, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36638713

ABSTRACT

BACKGROUND: The bark of Eucommia ulmoides (a perennial deciduous tree termed eucommia hereafter) has anti-hyperlipidemia effects due to its bioactive components. However, the slow growth of eucommia bark leads to a deficit in this resource. Studies have shown that eucommia leaf has bioactive components similar to those of eucommia bark and anti-hyperlipidemia effects. At present, the strength of the anti-hyperlipidemia effect of eucommia bark and eucommia leaf has not been reported. Their interaction with the gut microbiota and the mechanism by which the gut microbiota exerts anti-hyperlipidemia effects are unclear. PURPOSES: Through fecal microbiota transplantation (FMT) experiments, this study aimed to investigate the mechanism by which fecal bacteria suspensions containing chlorogenic acid (CGA), eucommia bark extract (EBE), and eucommia leaves extract (ELE) improve high-fat diet (HFD)-induced lipid metabolism disorders. Difference in anti-hyperlipidemia effects between EBE and ELE and exploring an eucommia bark substitute to improve the sustainable utilization of eucommia were also evaluated. RESULTS: EBE and ELE contain eight identical bioactive ingredients, and fecal bacteria suspensions containing EBE and ELE significantly improved HFD-induced lipid metabolism disorders and elevated blood glucose levels. The fecal bacteria suspension of healthy mice containing CGA, EBE, and ELE significantly reduced the relative abundance of Erysipelothrichaceae and Ruminococcaceae and promoted short chain fatty acids (SCFAs) production thereby activating the expression of the SCFA. G protein-coupled receptor 43 (GPR43) gene in colon and epididymal fat tissues. In addition, fecal bacteria suspensions of healthy mice containing CGA, EBE, or ELE significantly activated fasting-induced adipose factor (Fiaf) gene expression in colon tissue and inhibited the secretion of lipoprotein lipase (LPL) in liver tissue, thereby inhibiting the synthesis of triglycerides (TG). Changed in the Erysipelotrichaceae and Ruminococcaceae relative abundances were significantly correlated with these target genes. Thus, regulating the abundance of the Erysipelotrichaceae and Ruminococcaceae could serve as a potential target for the role of fecal bacteria suspensions of healthy mice containing CGA, EBE, or ELE in the Fiaf-LPL gut-liver axis and SCFAs-GPR43 gut-fat axis. In addition, regarding HFD-induced lipid metabolism disorders and gut microbiota structural disorders, we found no significant difference between ELE and EBE. CONCLUSIONS: Our FMT experiments evidenced that EBE and ELE improve lipid metabolism disorders by regulating the gut microbiota, providing a new pathway for treating hyperlipidemia using eucommia dietary therapy. There was no significant difference in the anti-hyperlipidemia effects of ELE and EBE; thus, eucommia leaf could replace eucommia bark in traditional Chinese medicine, so as to achieve a sustainable utilization of eucommia resources.


Subject(s)
Eucommiaceae , Gastrointestinal Microbiome , Lipid Metabolism Disorders , Mice , Animals , Diet, High-Fat/adverse effects , Lipid Metabolism , Eucommiaceae/chemistry , Lipoprotein Lipase , Plant Bark , Liver , Fatty Acids, Volatile/metabolism , Plant Extracts/therapeutic use , Lipid Metabolism Disorders/drug therapy , Lipid Metabolism Disorders/metabolism
3.
Front Oncol ; 11: 746917, 2021.
Article in English | MEDLINE | ID: mdl-34722304

ABSTRACT

As the main cause of death in the world, cancer is one of the major health threats for humans. In recent years, traditional Chinese medicine has gained great attention in oncology due to the features of multi-targets, multi-pathways, and slight side effects. Moreover, lots of traditional Chinese medicine can exert immunomodulatory effects in vivo. In the tumor microenvironment, tumor cells, immune cells as well as other stromal cells often coexist. With the development of cancer, tumor cells proliferate uncontrollably, metastasize aggressively, and modulate the proportion and status of immune cells to debilitate the antitumor immunity. Reversal of immunosuppressive tumor microenvironment plays an essential role in cancer prevention and therapy. Immunotherapy has become the most promising strategy for cancer therapy. Chinese medicine compounds can stimulate the activation and function of immune cells, such as promoting the maturation of dendritic cells and inducing the differentiation of myeloid-derived suppressor cells to dendritic cells and macrophages. In the present review, we summarize and discuss the effects of Chinese medicine compounds on immune cells in the tumor microenvironment, including innate immune cells (dendritic cells, natural killer cells, macrophages, and myeloid-derived suppressor cells) and adaptive immune cells (CD4+/CD8+ T lymphocytes and regulatory T cells), and the various immunomodulatory roles of Chinese medicine compounds in cancer therapy such as improving tumor-derived inflammation, enhancing the immunity after surgery or chemotherapy, blocking the immune checkpoints, et al., aiming to provide more thoughts for the anti-tumor mechanisms and applications of Chinese medicine compounds in terms of tumor immunity.

4.
Clin Transl Med ; 11(7): e410, 2021 07.
Article in English | MEDLINE | ID: mdl-34323416

ABSTRACT

BACKGROUND: Liver fibrosis and fibrosis-related hepatocarcinogenesis are a rising cause for morbidity and death worldwide. Although transforming growth factor-ß (TGF-ß) is a critical mediator of chronic liver fibrosis, targeting TGF-ß isoforms and receptors lead to unacceptable side effect. This study was designed to explore the antifibrotic effect of Compound kushen injection (CKI), an approved traditional Chinese medicine formula, via a therapeutic strategy of rebalancing TGF-ß/Smad7 signaling. METHODS: A meta-analysis was performed to evaluate CKI intervention on viral hepatitis-induced fibrosis or cirrhosis in clinical randomized controlled trials (RCTs). Mice were given carbon tetrachloride (CCl4 ) injection or methionine-choline deficient (MCD) diet to induce liver fibrosis, followed by CKI treatment. We examined the expression of TGF-ß/Smad signaling and typical fibrosis-related genes in hepatic stellate cells (HSCs) and fibrotic liver tissues by qRT-PCR, Western blotting, RNA-seq, immunofluorescence, and immunohistochemistry. RESULTS: Based on meta-analysis results, CKI improved the liver function and relieved liver fibrosis among patients. In our preclinical studies by using two mouse models, CKI treatment demonstrated promising antifibrotic effects and postponed hepatocarcinogenesis with improved liver function and histopathologic features. Mechanistically, we found that CKI inhibited HSCs activation by stabilizing the interaction of Smad7/TGF-ßR1 to rebalance Smad2/Smad3 signaling, and subsequently decreased the extracellular matrix formation. Importantly, Smad7 depletion abolished the antifibrotic effect of CKI in vivo and in vitro. Moreover, matrine, oxymatrine, sophocarpine, and oxysophocarpine were identified as material basis responsible for the antifibrosis effect of CKI. CONCLUSIONS: Our results unveil the approach of CKI in rebalancing TGF-ß/Smad7 signaling in HSCs to protect against hepatic fibrosis and hepatocarcinogenesis in both preclinical and clinical studies. Our study suggests that CKI can be a candidate for treatment of hepatic fibrosis and related oncogenesis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carbon Tetrachloride/toxicity , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line , Drugs, Chinese Herbal/therapeutic use , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Medicine, Chinese Traditional , Meta-Analysis as Topic , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/metabolism , Smad7 Protein/antagonists & inhibitors , Smad7 Protein/genetics
5.
J Immunother Cancer ; 8(1)2020 03.
Article in English | MEDLINE | ID: mdl-32179631

ABSTRACT

BACKGROUND: There is an urgent need for effective treatments for hepatocellular carcinoma (HCC). Immunotherapy is promising especially when combined with traditional therapies. This study aimed to investigate the immunomodulatory function of an approved Chinese medicine formula, compound kushen injection (CKI), and its anti-HCC efficiency in combination with low-dose sorafenib. METHODS: Growth of two murine HCC cells was evaluated in an orthotopic model, a subcutaneous model, two postsurgical recurrence model, and a tumor rechallenge model with CKI and low-dose sorafenib combination treatment. In vivo macrophage or CD8+ T cell depletion and in vitro primary cell coculture models were used to determine the regulation of CKI on macrophages and CD8+ T cells. RESULTS: CKI significantly enhanced the anticancer activity of sorafenib at a subclinical dose with no obvious side effects. CKI and sorafenib combination treatment prevented the postsurgical recurrence and rechallenged tumor growth. Further, we showed that CKI activated proinflammatory responses and relieved immunosuppression of tumor-associated macrophages in the HCC microenvironment by triggering tumor necrosis factor receptor superfamily member 1 (TNFR1)-mediated NF-κB and p38 MAPK signaling cascades. CKI-primed macrophages significantly promoted the proliferation and the cytotoxic ability of CD8+ T cells and decreased the exhaustion, which subsequently resulted in apoptosis of HCC cells. CONCLUSIONS: CKI acts on macrophages and CD8+ T cells to reshape the immune microenvironment of HCC, which improves the therapeutic outcomes of low-dose sorafenib and avoids adverse chemotherapy effects. Our study shows that traditional Chinese medicines with immunomodulatory properties can potentiate chemotherapeutic drugs and provide a promising approach for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Synergism , Drugs, Chinese Herbal/pharmacology , Immunosuppression Therapy , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sorafenib/pharmacology , Tumor-Associated Macrophages/immunology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Drugs, Chinese Herbal/administration & dosage , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
6.
Cancer Lett ; 454: 14-25, 2019 07 10.
Article in English | MEDLINE | ID: mdl-30910587

ABSTRACT

Patients receiving sorafenib treatment for hepatocellular carcinoma (HCC) experience different treatment efficacy. Personalized sorafenib treatment should be achieved through the identification of predictors of therapeutic response. In the current study, we found that high UGT1A9 expression indicated better prognosis for HCC patients treated with sorafenib after surgery. In silico analysis predicted microRNA-200a/-183 as potential regulators of the UGT1A gene family via binding to the shared UGT1A9 3'-UTR. A significant inverse correlation between microRNA-200a/-183 and UGT1A9 mRNA level was observed in a panel of HCC specimens. Direct binding was further demonstrated by luciferase reporter gene vector carrying wild-type or binding site truncated UGT1A9 3'-UTR. MicroRNA-200a/-183 downregulated UGT1A9 expression in a dose-dependent manner and significantly reduced sorafenib ß-D-glucuronide formation in HCC cells. These data indicated that UGT1A9, under epigenetic regulation of microRNA-200a/-183, could predict patients who might benefit from adjuvant sorafenib treatment after surgery.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Glucuronosyltransferase/genetics , Liver Neoplasms/drug therapy , MicroRNAs/genetics , Sorafenib/pharmacology , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/metabolism , HEK293 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , UDP-Glucuronosyltransferase 1A9 , Xenograft Model Antitumor Assays
7.
EBioMedicine ; 22: 58-67, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28754304

ABSTRACT

Hepatocellular carcinoma (HCC) is a common malignant tumor in the digestive tract with limited therapeutic choices. Although sorafenib, an orally administered multikinase inhibitor, has produced survival benefits for patients with advanced HCC, favorable clinical outcomes are limited due to individual differences and resistance. The application of immunotherapy, a promising approach for HCC is urgently needed. Macrophage infiltration, mediated by the CCL2/CCR2 axis, is a potential immunotherapeutic target. Here, we report that a natural product from Abies georgei, named 747 and related in structure to kaempferol, exhibits sensitivity and selectivity as a CCR2 antagonist. The specificity of 747 on CCR2 was demonstrated via calcium flux, the binding domain of CCR2 was identified in an extracellular loop by chimera binding assay, and in vivo antagonistic activity of 747 was confirmed through a thioglycollate-induced peritonitis model. In animals, 747 elevated the number of CD8+ T cells in tumors via blocking tumor-infiltrating macrophage-mediated immunosuppression and inhibited orthotopic and subcutaneous tumor growth in a CD8+ T cell-dependent manner. Further, 747 enhanced the therapeutic efficacy of low-dose sorafenib without obvious toxicity, through elevating the numbers of intra-tumoral CD8+ T cells and increasing death of tumor cells. Thus, we have discovered a natural CCR2 antagonist and have provided a new perspective on development of this antagonist for treatment of HCC. In mouse models of HCC, 747 enhanced the tumor immunosuppressive microenvironment and potentiated the therapeutic effect of sorafenib, indicating that the combination of an immunomodulator with a chemotherapeutic drug could be a new approach for treating HCC.


Subject(s)
Abies/chemistry , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Macrophages/drug effects , Plant Extracts/administration & dosage , Receptors, CCR2/antagonists & inhibitors , Animals , Binding Sites , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Hep G2 Cells , Humans , Immunosuppression Therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Macrophages/pathology , Mice , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/therapeutic use , Plant Extracts/chemistry , Plant Extracts/pharmacology , Receptors, CCR2/chemistry , Sorafenib , THP-1 Cells , Tumor Microenvironment , Xenograft Model Antitumor Assays
8.
EBioMedicine ; 14: 44-54, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27939426

ABSTRACT

Artemisinin (ARS) and its derivatives, which are clinically used antimalarial agents, have shown antitumor activities. Their therapeutic potencies, however, are limited by their low solubility and poor bioavailability. Here, through a pharmacophore hybridization strategy, we synthesized ARS-drug conjugates, in which the marketed chemotherapeutic agents chlorambucil, melphalan, flutamide, aminoglutethimide, and doxifluridine, were separately bonded to Dihydroartemisinin (DHA) through various linkages. Of these, the artemisinin-melphalan conjugate, ARS4, exhibited most toxicity to human ovarian cancer cells but had low cytotoxicity to normal cells. ARS4 inhibited the growth and proliferation of ovarian cancer cells and resulted in S-phase arrest, apoptosis, and inhibition of migration; these effects were stronger than those of its parent drugs, DHA and melphalan. Furthermore, ARS4 modulated the expression of proteins involved in cell cycle progression, apoptosis, and the epithelial-mesenchymal transition (EMT). Moreover, in mice, ARS4 inhibited growth and intraperitoneal dissemination and metastasis of ovarian cancer cells without observable toxic effects. Our results provide a basis for development of the compound as a chemotherapeutic agent. RESEARCH IN CONTEXT: Artemisinin compounds have recently received attention as anticancer agents because of their clinical safety profiles and broad efficacy. However, their therapeutic potencies are limited by low solubility and poor bioavailability. Here, we report that ARS4, an artemisinin-melphalan conjugate, possesses marked in-vitro and in-vivo antitumor activity against ovarian cancer, the effects of which are stronger than those for its parent drugs, Dihydroartemisinin and melphalan. In mice, ARS4 inhibits localized growth of ovarian cancer cells and intraperitoneal dissemination and metastasis without appreciable host toxicity. Thus, for patients with ovarian cancer, ARS4 is a promising chemotherapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Artemisinins/administration & dosage , Artemisinins/adverse effects , Artemisinins/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drug Combinations , Drug Evaluation, Preclinical , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Mice , Neoplasm Metastasis , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Chemistry ; 21(48): 17415-21, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26458147

ABSTRACT

Mitochondria-targeting theranostic probes that enable the simultaneously reporting of and triggering of mitochondrial dysfunctions in cancer cells are highly attractive for cancer diagnosis and therapy. Three fluorescent mitochondria-targeting theranostic probes have been developed by linking a mitochondrial dye, coumarin-3-carboximide, with a widely used traditional Chinese medicine, artemisinin, to kill cancer cells. Fluorescence images showed that the designed coumarin-artemisinin conjugates localized mainly in mitochondria, leading to enhanced anticancer activities over artemisinin. High cytotoxicity against cancer cells correlated with the strong ability to accumulate in mitochondria, which could efficiently increase the intracellular reactive oxygen species level and induce cell apoptosis. This study highlights the potential of using mitochondria-targeting fluorophores to selectively trigger and directly visualize subcellular drug delivery in living cells.


Subject(s)
Artemisinins/pharmacology , Coumarins/pharmacology , Fluorescent Dyes/pharmacology , Mitochondria/drug effects , Neoplasms/drug therapy , Apoptosis/drug effects , Artemisinins/chemistry , Drug Delivery Systems , Humans , Reactive Oxygen Species , Theranostic Nanomedicine
10.
Acta Pharmacol Sin ; 34(8): 1101-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23708556

ABSTRACT

AIM: To investigate the embryotoxicity of dihydroartemisinin (DHA), the main active metabolite of artemisinin, in zebrafish, and explore the corresponding mechanisms. METHODS: The embryos of wild type and TG (flk1:GFP) transgenic zebrafish were exposed to DHA. Developmental phenotypes of the embryos were observed. Development of blood vessels was directly observed in living embryos of TG (flk1:GFP) transgenic zebrafish under fluorescence microscope. The expression of angiogenesis marker genes vegfa, flk1, and flt1 in the embryos was detected using real-time PCR and RNA in situ hybridization assays. RESULTS: Exposure to DHA (1-10 mg/L) dose-dependently caused abnormal zebrafish embryonic phenotypes in the early developmental stage. Furthermore, exposure to DHA (10 mg/L) resulted in more pronounced embryonic angiogenesis in TG (flk1:GFP) zebrafish line. Exposure to DHA (10 mg/L) significantly increased the mRNA expression of vegfa, flk1, and flt1 in the embryos. Knockdown of the flk1 protein partially blocked the effects of DHA on embryogenesis. CONCLUSION: DHA causes abnormal embryonic phenotypes and promotes angiogenesis in zebrafish early embryonic development, demonstrating the potential embryotoxicity of DHA.


Subject(s)
Artemisia/toxicity , Artemisinins/toxicity , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Neovascularization, Pathologic/chemically induced , Zebrafish/embryology , Animals , Animals, Genetically Modified , Embryo, Nonmammalian/pathology , Embryonic Development/drug effects , Embryonic Development/physiology , Neovascularization, Pathologic/pathology , Zebrafish/genetics
11.
Curr Cancer Drug Targets ; 13(4): 472-83, 2013 May.
Article in English | MEDLINE | ID: mdl-23597199

ABSTRACT

Identification of novel chemotherapeutic agents from traditional medicines and elucidation of the molecular basis of their anticancer effects are critical and urgently needed for modern pharmacotherapy. We previously found that analogs of the compounds present in Valeriana jatamansi, a traditional medicine used to treat mental disorders, possess notable antitumor properties; however, the underlying molecular mechanisms have not been fully demonstrated. In this study, we evaluated the anticancer effects of IVHD-valtrate, one of the most active Valeriana jatamansi derivatives, against human ovarian cancer cells in vitro and in vivo. IVHD-valtrate inhibited the growth and proliferation of the A2780 and OVCAR-3 ovarian cancer cell lines in a concentration-dependent manner, while relatively low cytotoxicity to immortalized non-tumorigenic human ovarian surface epithelial cells (IOSE-144) was observed. Treatment with IVHD-valtrate arrested the ovarian cancer cells in the G2/M phase and induced apoptosis, and significantly suppressed the growth of A2780 and OVCAR3 xenograft tumors in a dose-dependent manner. The detailed in vitro and in vivo study on the molecular mechanisms of this compound demonstrated that IVHD-valtrate exposure modulated the expression of numerous molecules involved in cell cycle progression and apoptosis regardless of p53 status, leading to increase the level of p53, Rb, p21, p27 and decrease Mdm2, E2F1, Cyclin B1, Cdc25C and Cdc2. It also down-regulated Bcl-2/Bax and Bcl-2/Bad ratio and enhanced the cleavage of PARP and Caspases. Our preclinical results indicated IVHD-valtrate is a potential therapeutic agent for ovarian cancer, providing a basis for development of the compound as a novel chemotherapeutic agent.


Subject(s)
Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Iridoids/pharmacology , Ovarian Neoplasms/drug therapy , Phytotherapy , Plant Extracts/pharmacology , Valerian/chemistry , Animals , Biomarkers, Tumor/genetics , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
12.
Drug Metab Pharmacokinet ; 26(2): 201-5, 2011.
Article in English | MEDLINE | ID: mdl-21178302

ABSTRACT

Ginseng is one of the most commonly used herbal medicines worldwide. Ginsenosides are believed to be responsible for the therapeutic activities of ginseng; however, co-administration of prescription drugs with ginseng products may give rise to ginseng-drug interactions. Cytochrome P450 enzymes are major phase I enzymes involved in the metabolism of most currently used drugs. Inhibition or induction of P450 enzymes can lead to pharmacokinetic drug interactions. Previous reports on ginseng-drug interactions have been controversial and confusing. In the present study, we examined the effects of thirteen ginsenosides on the expression of CYP1A1, CYP1A2 and CYP3A4 in HepG2 cells. We found that eight ginsenosides and aglycones potently induced CYP1A1 expression, and that structure-activity relationships existed for these effects. Moreover, we discovered that deglycosylated ginsenosides, some of which are putative ginsenoside metabolites, were more potent inducers of CYP1A1, CYP1A2 and CYP3A4 than glycosylated ginsenosides. This finding indicates that ginsenoside metabolites may partially account for ginseng-drug interactions, and that differences in the composition of intestinal bacteria and the extent of deglycosylation of the ginsenosides could be a contributing factor to the inconsistencies observed in previous clinical and pre-clinical studies with regard to ginseng-drug interactions.


Subject(s)
Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A2/drug effects , Cytochrome P-450 CYP3A/drug effects , Enzyme Induction/drug effects , Ginsenosides , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Ginsenosides/chemistry , Ginsenosides/metabolism , Ginsenosides/pharmacology , Glycosylation , Hep G2 Cells , Herb-Drug Interactions , Humans , Panax/metabolism , Plant Extracts/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL