Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Zhongguo Zhong Yao Za Zhi ; 48(24): 6663-6675, 2023 Dec.
Article in Chinese | MEDLINE | ID: mdl-38212026

ABSTRACT

The study investigated the effects of different processed products of Polygonati Rhizoma(black bean-processed Polygonati Rhizoma, BBPR; stewed Polygonati Rhizoma, SPR) on the urinary metabolites in a rat model of Alzheimer's disease(AD). Sixty SPF-grade male SD rats were randomized into a control group, a model group, a donepezil group, a BBPR group, and a SPR group, with twelve rats in each group. Other groups except the control group were administrated with D-galactose injection(100 mg·kg~(-1)) once a day for seven weeks. The control group was administrated with an equal volume of normal saline once a day for seven consecutive weeks. After three weeks of D-galactose injection, bilateral hippocampal Aß_(25-35) injections were performed for modeling. The rats were administrated with corresponding drugs(10 mL·kg~(-1)) by gavage since week 2, and the rats in the model and control group with an equal volume of double distilled water once a day for 35 continuous days. The memory behaviour and pathological changes in the hippocampal tissue were observed. The untargeted metabolites in the urine were detected by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry(UPLC-Q/TOF-MS). Principal component analysis(PCA) and orthogonal partial least square-discriminant analysis(OPLS-DA) were employed to characterize and screen differential metabolites and potential biomarkers, for which the metabolic pathway enrichment analysis was conducted. The results indicated that BBPR and SPR increased the new object recognition index, shortened the escape latency, and increased the times of crossing the platform of AD rats in the Morris water maze test. The results of hematoxylin-eosin(HE) staining showed that the cells in the hippocampal tissue of the drug administration groups were closely arranged. Moreover, the drugs reduced the content of interleukin-6(IL-6, P<0.01) and tumor necrosis factor-α(TNF-α) in the hippocampal tissue, which were more obvious in the BBPR group(P<0.05). After screening, 15 potential biomarkers were identified, involving two metabolic pathways: dicoumarol pathway and piroxicam pathway. BBPR and SPR may alleviate AD by regulating the metabolism of dicoumarol and piroxicam.


Subject(s)
Alzheimer Disease , Rats , Male , Animals , Alzheimer Disease/drug therapy , Chromatography, High Pressure Liquid/methods , Rats, Sprague-Dawley , Dicumarol , Galactose , Piroxicam , Metabolomics/methods , Biomarkers/urine
2.
Article in Chinese | MEDLINE | ID: mdl-36229221

ABSTRACT

Bromadiolone, commonly known as super warfarin, is a long-acting coumarin dicoumarin rodenticide. The mechanism of bromadiolone is mainly to inhibit vitamin K1 epoxide reductase and affect the synthesis of coagulation factors Ⅱ, Ⅶ, Ⅸ and Ⅹ, which causes blood coagulation dysfunction and systemic multiple organ hemorrhage. Here, we report of a case of bromadiolone poisoning patient who had digestive tract, abdominal hemorrhage, as well as secondary paralytic ileus. After blood product transfusion and vitamin K1 supplementation, the patient was discharged after the physical condition was improved. It's suggestied that clinicians should pay attention to rare complications to prevent missed diagnosis when treating other bromadiolone poisoning.


Subject(s)
4-Hydroxycoumarins , Intestinal Pseudo-Obstruction , Rodenticides , Blood Coagulation Factors , Dicumarol , Hemorrhage , Humans , Intestinal Pseudo-Obstruction/chemically induced , Oxidoreductases , Vitamin K 1 , Warfarin
3.
Molecules ; 27(7)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35408682

ABSTRACT

A new dicoumarin, jusan coumarin, (1), has been isolated from Artemisia glauca aerial parts. The chemical structure of jusan coumarin was estimated, by 1D, 2D NMR as well as HR-Ms spectroscopic methods, to be 7-hydroxy-6-methoxy-3-[(2-oxo-2H-chromen-6-yl)oxy]-2H-chromen-2-one. As the first time to be introduced in nature, its potential against SARS-CoV-2 has been estimated using various in silico methods. Molecular similarity and fingerprints experiments have been utilized for 1 against nine co-crystallized ligands of COVID-19 vital proteins. The results declared a great similarity between Jusan Coumarin and X77, the ligand of COVID-19 main protease (PDB ID: 6W63), Mpro. To authenticate the obtained outputs, a DFT experiment was achieved to confirm the similarity of X77 and 1. Consequently, 1 was docked against Mpro. The results clarified that 1 bonded in a correct way inside Mpro active site, with a binding energy of -18.45 kcal/mol. Furthermore, the ADMET and toxicity profiles of 1 were evaluated and showed the safety of 1 and its likeness to be a drug. Finally, to confirm the binding and understand the thermodynamic characters between 1 and Mpro, several molecular dynamics (MD) simulations studies have been administered. Additionally, the known coumarin derivative, 7-isopentenyloxycoumarin (2), has been isolated as well as ß-sitosterol (3).


Subject(s)
Artemisia , Coronavirus 3C Proteases , Coumarins , Protease Inhibitors , SARS-CoV-2 , Artemisia/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coumarins/chemistry , Coumarins/pharmacology , Dicumarol/chemistry , Dicumarol/pharmacology , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology
4.
PLoS One ; 14(2): e0212233, 2019.
Article in English | MEDLINE | ID: mdl-30779774

ABSTRACT

Currently, there is no available therapy to eradicate hepatitis B virus (HBV) in chronically infected individuals. This is due to the difficulty in eliminating viral covalently closed circular (ccc) DNA, which is central to the gene expression and replication of HBV. We developed an assay system for nuclear circular DNA using an integration-deficient lentiviral vector. This vector produced non-integrated circular DNA in nuclei of infected cells. We engineered this vector to encode firefly luciferase to monitor the lentiviral episome DNA. We screened 3,840 chemicals by this assay for luciferase-reducing activity and identified dicumarol, which is known to have anticoagulation activity. We confirmed that dicumarol reduced lentiviral episome DNA. Furthermore, dicumarol inhibited HBV replication in cell culture using NTCP-expressing HepG2 and primary human hepatocytes. Dicumarol reduced intracellular HBV RNA, DNA, supernatant HBV antigens and DNA. We also found that dicumarol reduced the cccDNA level in HBV infected cells, but did not affect HBV adsorption/entry. This is a novel assay system for screening inhibitors targeting nuclear cccDNA and is useful for finding new antiviral substances for HBV.


Subject(s)
Antiviral Agents/pharmacology , Cell Nucleus/metabolism , DNA, Viral/metabolism , Dicumarol/pharmacology , Hepatitis B virus/metabolism , Plasmids/metabolism , Cell Nucleus/genetics , Cell Nucleus/virology , DNA, Viral/genetics , Drug Evaluation, Preclinical , Genetic Vectors , HEK293 Cells , Hep G2 Cells , Hepatitis B virus/genetics , Humans , Lentivirus , Plasmids/genetics , RNA, Viral/genetics , RNA, Viral/metabolism
5.
Sci Rep ; 6: 27566, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27297123

ABSTRACT

UNLABELLED: Chemical regulation of macrophage function is one key strategy for developing host-directed adjuvant therapies for tuberculosis (TB). A critical step to develop these therapies is the identification and characterization of specific macrophage molecules and pathways with a high potential to serve as drug targets. Using a barcoded lentivirus-based pooled short-hairpin RNA (shRNA) library combined with next generation sequencing, we identified 205 silenced host genes highly enriched in mycobacteria-resistant macrophages. Twenty-one of these "hits" belonged to the oxidoreductase functional category. NAD(P)H: quinone oxidoreductase 1 (NQO1) was the top oxidoreductase "hit". NQO1 expression was increased after mycobacterial infection, and NQO1 knockdown increased macrophage differentiation, NF-κB activation, and the secretion of pro-inflammatory cytokines TNF-α and IL-1ß in response to infection. This suggests that mycobacteria hijacks NQO1 to down-regulate pro-inflammatory and anti-bacterial functions. The competitive inhibitor of NQO1 dicoumarol synergized with rifampin to promote intracellular killing of mycobacteria. Thus, NQO1 is a new host target in mycobacterial infection that could potentially be exploited to increase antibiotic efficacy in vivo. Our findings also suggest that pooled shRNA libraries could be valuable tools for genome-wide screening in the search for novel druggable host targets for adjunctive TB therapies.


Subject(s)
Antitubercular Agents/pharmacology , Dicumarol/pharmacology , Host-Pathogen Interactions/drug effects , Macrophages/immunology , Mycobacterium tuberculosis/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , Drug Synergism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Gene Library , High-Throughput Nucleotide Sequencing , High-Throughput Screening Assays , Humans , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/physiology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/immunology , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rifampin/pharmacology , Signal Transduction , THP-1 Cells , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Bull Exp Biol Med ; 159(1): 44-7, 2015 May.
Article in English | MEDLINE | ID: mdl-26033588

ABSTRACT

Cell damage depending on activity of quinone reductase 2 (MT3 receptor) was simulated in experiments on bone marrow cell suspension and assessed by menadione-induced DNA breaks measured by comet assay. We analyzed the protective effect of afobazole interacting with MT1, MT3, σ1 receptors, and monoamine oxidase A and its main metabolite M11 that specifi cally binds to MT3 receptors. Both compounds reduced the level of menadione-induced DNA damage (afobazole was effective in lower concentrations in comparison with M-11). Conclusion was made on the contribution of MT3 receptors to the protective effect of afobazole, but the observed concentration differences indicate possible contribution of other targets of anxiolytic drug to the protective mechanisms.


Subject(s)
Anti-Anxiety Agents/pharmacology , Benzimidazoles/pharmacology , Bone Marrow Cells/drug effects , DNA Breaks/drug effects , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Quinone Reductases/antagonists & inhibitors , Receptors, Melatonin/drug effects , Animals , Anti-Anxiety Agents/metabolism , Benzimidazoles/metabolism , Biotransformation , Cells, Cultured , Comet Assay , Dicumarol/pharmacology , Drug Evaluation, Preclinical , Metallothionein 3 , Mice , Monoamine Oxidase , Monoamine Oxidase Inhibitors , Morpholines/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Neuroprotective Agents/metabolism , Quinone Reductases/metabolism , Receptor, Melatonin, MT1/drug effects , Receptors, sigma/drug effects , Vitamin K 3/toxicity
7.
PLoS One ; 9(7): e101959, 2014.
Article in English | MEDLINE | ID: mdl-25051199

ABSTRACT

AIMS: Chewing of betel quid (BQ) increases the risk of oral cancer and oral submucous fibrosis (OSF), possibly by BQ-induced toxicity and induction of inflammatory response in oral mucosa. METHODS: Primary gingival keratinocytes (GK cells) were exposed to areca nut (AN) components with/without inhibitors. Cytotoxicity was measured by 3-(4,5-dimethyl- thiazol- 2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. mRNA and protein expression was evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR) and western blotting. PGE2/PGF2α production was measured by enzyme-linked immunosorbent assays. RESULTS: Areca nut extract (ANE) stimulated PGE2/PGF2α production, and upregulated the expression of cyclooxygenase-2 (COX-2), cytochrome P450 1A1 (CYP1A1) and hemeoxygenase-1 (HO-1), but inhibited expression of keratin 5/14, cyclinB1 and cdc25C in GK cells. ANE also activated epidermal growth factor receptor (EGFR), Src and Ras signaling pathways. ANE-induced COX-2, keratin 5, keratin 14 and cdc25C expression as well as PGE2 production were differentially regulated by α-naphthoflavone (a CYP 1A1/1A2 inhibitor), PD153035 (EGFR inhibitor), pp2 (Src inhibitor), and manumycin A (a Ras inhibitor). ANE-induced PGE2 production was suppressed by piper betle leaf (PBL) extract and hydroxychavicol (two major BQ components), dicoumarol (a NAD(P)H: Quinone Oxidoreductase--NQO1 inhibitor) and curcumin. ANE-induced cytotoxicity was inhibited by catalase and enhanced by dicoumarol, suggesting that AN components may contribute to the pathogenesis of OSF and oral cancer via induction of aberrant differentiation, cytotoxicity, COX-2 expression, and PGE2/PGF2α production. CONCLUSIONS: CYP4501A1, reactive oxygen species (ROS), EGFR, Src and Ras signaling pathways could all play a role in ANE-induced pathogenesis of oral cancer. Addition of PBL into BQ and curcumin consumption could inhibit the ANE-induced inflammatory response.


Subject(s)
Areca/chemistry , Gene Expression/drug effects , Keratinocytes/metabolism , Plant Extracts/toxicity , Cells, Cultured , Curcumin/pharmacology , Cyclin B1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 CYP1A1/metabolism , Dicumarol/pharmacology , Dinoprostone/biosynthesis , ErbB Receptors/metabolism , Gingiva/pathology , Heme Oxygenase-1/metabolism , Humans , Keratinocytes/drug effects , Keratins/genetics , Keratins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , ras Proteins/metabolism , src-Family Kinases/metabolism
8.
J Plant Physiol ; 167(11): 870-8, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20181408

ABSTRACT

Hyoscyamus albus hairy roots secrete riboflavin under Fe-deficient conditions. To determine whether this secretion was linked to an enhancement of respiration, both riboflavin secretion and the reduction of 2,3,5-triphenyltetrazolium chloride (TTC), as a measure of respiration activity, were determined in hairy roots cultured under Fe-deficient and Fe-replete conditions, with or without aeration. Appreciable TTC-reducing activity was detected at the root tips, at the bases of lateral roots and in internal tissues, notably the vascular system. TTC-reducing activity increased under Fe deficiency and this increase occurred in concert with riboflavin secretion and was more apparent under aeration. Riboflavin secretion was not apparent under Fe-replete conditions. In order to examine which elements of the mitochondrial electron transport chain might be involved, the effects of the respiratory inhibitors, barbiturate, dicoumarol, malonic acid, antimycin, KCN and salicylhydroxamic acid (SHAM) were investigated. Under Fe-deficient conditions, malonic acid affected neither root growth, TTC-reducing activity nor riboflavin secretion, whereas barbiturate and SHAM inhibited only root growth and TTC-reducing activity, respectively, and the other compounds variously inhibited growth and TTC-reducing activity. Riboflavin secretion was decreased, in concert with TTC-reducing activity, by dicoumarol, antimycin and KCN, but not by SHAM. In Fe-replete roots, all inhibitors which reduced riboflavin secretion in Fe-deficient roots showed somewhat different effects: notably, antimycin and KCN did not significantly inhibit TTC-reducing activity and the inhibition by dicoumarol was much weaker in Fe-replete roots. Combined treatment with KCN and SHAM also revealed that Fe-deficient and Fe-replete roots reduced TTC in different ways. A decrease in the Fe content of mitochondria in Fe-deficient roots was confirmed. Overall, the results suggest that, under conditions of Fe deficiency in H. albus hairy roots, the alternative NAD(P)H dehydrogenases, complex III and complex IV, but not the alternative oxidase, are actively involved both in respiration and in riboflavin secretion.


Subject(s)
Electron Transport/physiology , Hyoscyamus/metabolism , Iron Deficiencies , Plant Roots/metabolism , Riboflavin/metabolism , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Barbiturates/pharmacology , Cyanates/pharmacology , Dicumarol/pharmacology , Electron Transport/drug effects , Hyoscyamus/drug effects , Malonates/pharmacology , Plant Roots/drug effects , Salicylamides/pharmacology
9.
Fitoterapia ; 80(5): 283-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19306914

ABSTRACT

A new dicoumarin, named as dimeresculetin (1), together with another dicoumarin, euphorbetin (2) and esculetin (3) were isolated from the ethyl acetate extract of the dried whole plants of Viola yedoensis Makino. The structure of 1 was elucidated as 7-hydroxy-6-[(6,7-dihydroxy-2-oxo-2H-1-benzopyran-5-yl)oxy]-2H-1-benzopyran-2-one on the basis of extensive NMR, as well as the other spectral analysis. Compounds 1-3 exhibited anticoagulant activities with respect to activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT).


Subject(s)
Anticoagulants/isolation & purification , Coumarins/isolation & purification , Dicumarol/isolation & purification , Plant Extracts/isolation & purification , Umbelliferones/isolation & purification , Viola/chemistry , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Coumarins/chemistry , Dicumarol/chemistry , Dicumarol/pharmacology , Heparin/pharmacology , Partial Thromboplastin Time , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Prothrombin Time , Rabbits , Thrombin Time , Umbelliferones/chemistry , Umbelliferones/pharmacology
10.
Curr Drug Metab ; 8(2): 137-49, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17305492

ABSTRACT

Quinones represent a very important class of compounds found in nature and for the chemically synthesized drugs. The present study was designed to elucidate the intestinal first pass metabolic pathways in vivo and in vitro, of tanshinone IIA (TS), a derivative of phenanthrene-quinone isolated from Salvia miltiorrhiza. Five metabolites, proposed to be TS catechol glucuronides (two position isomers), dehydrotanshinone IIA and its two catechol glucuronides, were identified from the rat intestinal homogenates after oral administration of TS. TS metabolism was further conducted in the subcellular system including cytosol, microsomes, mitochondrial and S9 under both phase I and phase II metabolic conditions. TS underwent negligible metabolism in all of the subcellular systems under phase I metabolic condition using NADPH as the cofactor. However, significant and substantial metabolic elimination of TS was observed in the cytosol and S9 fractions, while not in the microsomes fractions, when both NADPH and UDPGA were added. Two TS catechol glucuronides were identified from such an in vitro metabolic medium. Dicoumarol, a specific inhibitor of the NAD(P)H dependent quinone oxidoreductase (NQO1), significantly inhibited the metabolic elimination of TS in a noncompetitive way, suggesting that NQO1 was responsible for the quinone reduction of TS to form the catechol intermediate. The catechol intermediate failed to be detected directly was proved to be highly unstable and autoxidized back to TS accompanied with hydrogen peroxide generation. Dicoumarol exhibited a significant inhibitory effect on the hydrogen peroxide generation, further supporting that the reduction of TS was catalyzed by NQO1. The absolute bioavailability of TS was significantly enhanced by oral dicoumarol pretreatment. In conclusion, a novel intestinal metabolic pathway for quinones, NQO1 mediated reduction and subsequent glucuronidation, was determined using TS as a model compound. This study should be helpful for the general understanding of quinones absorption and intestinal first pass metabolism.


Subject(s)
Drugs, Chinese Herbal/pharmacokinetics , Intestinal Mucosa/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phenanthrenes/pharmacokinetics , Quinones/pharmacokinetics , Abietanes , Animals , Dicumarol/pharmacology , Drugs, Chinese Herbal/administration & dosage , Glucuronides/metabolism , Intestines/drug effects , Liver/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Oxidation-Reduction , Phenanthrenes/administration & dosage , Quinones/administration & dosage , Rats , Rats, Sprague-Dawley
11.
Clin Cancer Res ; 11(24 Pt 1): 8866-71, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16361576

ABSTRACT

PURPOSE: The purpose of the present study was to evaluate the efficacy of mild hyperthermia to potentiate the anticancer effects of beta-lapachone (3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione) by up-regulating NAD(P)H:quinone oxidoreductase (NQO1) in cancer cells. EXPERIMENTAL DESIGN: Effects of beta-lapachone alone or in combination with mild heating on the clonogenic survival of FSaII fibrosarcoma cells of C3H mice and A549 human lung tumor cells in vitro was determined. Effects of heating on the NQO1 level in the cancer cells in vitro were assessed using Western blot analysis for NQO1 expression, biochemical determination of NQO1 activity, and immunofluorescence microscopy for NQO1 expression. Growth of FSaII tumors in the hind legs of C3H mice was determined after treating the host mice with i.p. injection of 45 mg/kg beta-lapachone followed by heating the tumors at 42 degrees C for 1 hour every other day for four times. RESULTS: Incubation of FSaII tumor cells and A549 tumor cells with beta-lapachone at 37 degrees C reduced clonogenic survival of the cells in dose-dependent and incubation time-dependent manner. NQO1 level in the cancer cells in vitro increased within 1 hour after heating at 42 degrees C for 1 hour and remained elevated for >72 hours. The clonogenic cell death caused by beta-lapachone increased in parallel with the increase in NQO1 levels in heated cells. Heating FSaII tumors in the legs of C3H mice enhanced the effect of i.p.-injected beta-lapachone in suppressing tumor growth. CONCLUSION: We observed for the first time that mild heat shock up-regulates NQO1 in tumor cells. The heat-induced up-regulation of NQO1 enhanced the anticancer effects of beta-lapachone in vitro and in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Hyperthermia, Induced , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/therapeutic use , Neoplasms/therapy , Animals , Cell Death , Cell Line, Tumor , Combined Modality Therapy , Dicumarol/therapeutic use , Enzyme Inhibitors/pharmacology , Humans , Mice , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Up-Regulation
12.
Chem Res Toxicol ; 18(8): 1296-305, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16097803

ABSTRACT

The female parts of hops (Humulus lupulus L.) show estrogenic effects as well as cancer chemopreventive potential. We analyzed the chemopreventive mechanism of hops by studying its antioxidative activities and its effect on the detoxification of a potentially toxic quinone (menadione). The detoxification enzyme quinone reductase [(NAD(P)H:quinone oxidoreductase, QR] protects against quinone-induced toxicity and has been used as a marker in cancer chemoprevention studies. Although the hop extract was only a weak quencher of free radicals formed from 1,1-diphenyl-2-picrylhydrazyl, it demonstrated strong QR induction in Hepa 1c1c7 cells. In addition, compounds isolated from hops including xanthohumol (XH) and 8-prenylnaringenin were tested for QR induction. Among these, XH was the most effective at inducing QR with a concentration required to double the specific activity of QR (CD value) of 1.7 +/- 0.7 microM. In addition, pretreatment of Hepa1c1c7 cells with XH significantly inhibited menadione-induced DNA single-strand breaks. The QR inhibitor dicumarol reversed the protective effect of XH against menadione-induced DNA damage. Because the expression of QR and other detoxifying enzymes is known to be upregulated by binding of the transcription factor Nrf2 to the antioxidant response element (ARE), the reporter activity mediated by ARE in HepG2-ARE-C8 cells was investigated after incubation with XH for 24 h. Under these conditions, XH increased ARE reporter activity in a dose-dependent manner. One mechanism by which XH might induce QR could be through interaction with Keap1, which sequesters Nrf2 in the cytoplasm, so that it cannot activate the ARE. Using LC-MS-MS, we demonstrated that XH alkylates human Keap1 protein, most likely on a subset of the 27 cysteines of Keap1. This suggests that XH induces QR by covalently modifying the Keap1 protein. Therefore, XH and hops dietary supplements might function as chemopreventive agents, through induction of detoxification enzymes such as QR.


Subject(s)
DNA Damage/drug effects , Humulus/chemistry , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Propiophenones/pharmacology , Vitamin K 3/antagonists & inhibitors , Vitamin K 3/toxicity , Adaptor Proteins, Signal Transducing/metabolism , Alkylation , Animals , Antioxidants/metabolism , Biphenyl Compounds/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoskeletal Proteins/metabolism , Dicumarol/pharmacology , Enzyme Induction/drug effects , Flavonoids , Genes, Reporter/drug effects , Hydrazines/metabolism , Kelch-Like ECH-Associated Protein 1 , Luciferases/metabolism , Mice , Picrates , Plant Extracts/pharmacology , Propiophenones/isolation & purification , Protein Prenylation , Response Elements , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
J Biomed Sci ; 12(2): 347-61, 2005.
Article in English | MEDLINE | ID: mdl-15917998

ABSTRACT

Tanshinone IIA, a major component extracted from the traditional herbal medicine, Salvia miltiorrhiza Bunge, is known to exhibit potent cytotoxicity against various human carcinoma cells in vitro. However, the mechanism by which tanshinone IIA produces this anti-tumor effect remains unknown. Since anti-neovascularization has generally been regarded as an effective strategy for anti-cancer therapy, we decided to investigate the mechanism underlying tanshinone IIA-mediated death of human endothelial cells. In this study, we demonstrate that tanshinone IIA elicits human endothelial cell death independent of oxidative stress. These events are partially calcium-dependent and actually dependent upon NAD(P)H: quinone oxidoreductase (NQO1) activity. Tanshinone IIA induces an increase in intracellular calcium, which triggers the release of cytochrome c, thus causing loss of the mitochondrial membrane potential (MMP), resulting in the subsequent activation of caspases. Blocking the induction of Ca2+ perturbation with BAPTA-AM partially rescued cells from tanshinone IIA-induced cytotoxicity. Additionally, blocking NQO1 activity with dicoumoral or inhibiting caspase activities with the general caspase inhibitor, z-VAD-fmk, prevented cell death induced by tanshinone IIA. Therefore, our results imply that tanshinone IIA-mediated cytotoxicity against human endothelial cells may occur through activation of NQO1, which induces a calcium imbalance and mitochondrial dysfunction, thus stimulating caspase activity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Endothelial Cells/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phenanthrenes/pharmacology , Salvia miltiorrhiza/metabolism , Abietanes , Acridine Orange/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Calcium/metabolism , Caspase Inhibitors , Caspases/metabolism , Cell Cycle , Cell Death , Cytochromes c/metabolism , Dicumarol/pharmacology , Drugs, Chinese Herbal , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/pathology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Membrane Potentials , Mitochondria/metabolism , Mitochondria/pathology , Models, Biological , Oxidative Stress , Plant Extracts , Reactive Oxygen Species/metabolism , Time Factors
14.
Chem Res Toxicol ; 18(3): 415-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777081

ABSTRACT

We report a new and specific mechanism for iron-mediated neurotoxicity using RCHT cells, which were derived from rat hypothalamus. RCHT cells exhibit immunofluorescent-positive markers for dopamine beta-hydroxylase and the norepinephrine transporter, NET. In the present study, we observed that iron-induced neurotoxicity in RCHT cells was dependent on (i) formation of an Fe-dopamine complex (100 microM FeCl3:100 microM dopamine); (ii) specific uptake of the Fe-dopamine complex into RCHT cells via NET (79+/-2 pmol 59Fe/mg/min; P<0.05), since the uptake of the 59Fe-dopamine complex by the cells was inhibited by 30 microM reboxetine, a specific NET inhibitor (78% inhibition, P<0.001); and (iii) intracellular oxidation of dopamine present in the Fe-dopamine complex to aminochrome; (iv) inhibition of DT-diaphorase, since incubation of RCHT cells with 100 microM Fe-dopamine complex in the presence of 100 microM dicoumarol, an inhibitor of DT-diaphorase, induced significant cell death (51+/-5%; P<0.001). However, this cell death was reduced by 75% when the cells were incubated in the presence of 30 microM reboxetine (P<0.01). No significant cell death was observed when the cells were incubated with 100 microM dopamine, 100 microM Fe-Dopamine complex, 100 microM dicoumarol, or 100 microM FeCl3 (8.3+/-2, 9+/-4, 8.5+/-3, or 9.7+/-2% of control, respectively). ESR studies using the spin trapping agent DMPO showed no formation of hydroxyl radicals when the cells were incubated with 100 microM FeCl3 alone. However, using the same ESR technique, the formation of hydroxyl radicals and a carbon-centered radical was detected when the cells were incubated with 100 microM Fe-dopamine complex in the presence of 100 microM dicoumarol. These studies suggest that iron can induce cell toxicity by a mechanism that requires the formation and NET-mediated uptake of an Fe-dopamine complex, ultimately resulting in the intracellular formation of reactive species.


Subject(s)
Dopamine/metabolism , Ferric Compounds/metabolism , Hypothalamus/drug effects , Iron/toxicity , Adrenergic Uptake Inhibitors/pharmacology , Animals , Cell Death/drug effects , Cell Line , Chlorides , Dicumarol/pharmacology , Dopamine/pharmacology , Dopamine beta-Hydroxylase/metabolism , Fluorescent Antibody Technique , Hypothalamus/enzymology , Hypothalamus/pathology , Indolequinones/metabolism , Iron/metabolism , Microscopy, Confocal , Morpholines/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins , Rats , Rats, Inbred F344 , Reboxetine , Symporters/metabolism
15.
Free Radic Res ; 38(11): 1223-33, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15621700

ABSTRACT

Manganese superoxide dismutase (MnSOD) levels have been found to be low in human pancreatic cancer [Pancreas 26, (2003), 23] and human pancreatic cancer cell lines [Cancer Res. 63, (2003), 1297] when compared to normal human pancreas. We hypothesized that stable overexpression of pancreatic cancer cells with MnSOD cDNA would alter the malignant phenotype. MIA PaCa-2 cells were stably transfected with a pcDNA3 plasmid containing sense human MnSOD cDNA or containing no MnSOD insert by using the lipofectAMINE method. G418-resistant colonies were isolated, grown and maintained. Overexpression of MnSOD was confirmed in two selected clones with a 2-4-fold increase in MnSOD immunoreactive protein. Compared with the parental and neo control cells, the MnSOD-overexpressing clones had decreased growth rates, growth in soft agar and plating efficiency in vitro, while in vivo, the MnSOD-overexpressing clones had slower growth in nude mice. These results suggest that MnSOD may be a tumor suppressor gene in human pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Agar , Aged , Animals , Antioxidants/metabolism , Cell Proliferation , Cells, Cultured , DNA, Complementary/genetics , Dicumarol/pharmacology , Gene Expression/radiation effects , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Superoxides/metabolism , Transfection
16.
Chem Pharm Bull (Tokyo) ; 52(11): 1362-4, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15516764

ABSTRACT

Two new type dicoumarins (dicoumaro-p-menthanes), named dibothrioclinins I (1) and II (2) were isolated from the roots and rhizomes of Gerbera piloselloides (L.) CASS., collected in Yunnan Province, China. Their structures were elucidated on the basis of MS, 1D ((1)H-NMR, (13)C-NMR, DEPT and NOE) and 2D ((1)H-(1)H COSY, HMQC, HMBC) NMR spectral analyses. The relative structures of the two compounds were established by NOE difference spectroscopy and further confirmed by single-crystal X-ray diffraction studies.


Subject(s)
Asteraceae , Dicumarol/chemistry , Dicumarol/isolation & purification , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Plant Roots , Rhizome
17.
Life Sci ; 75(13): 1649-57, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15261768

ABSTRACT

Gastrodia elata (G. elata) is a traditional Chinese herbal medicine for treating headaches, dizziness, tetanus, and epilepsy. In this study, differential methanol (MeOH) extracts of G. elata were found to prevent serum-deprived rat pheochromocytoma (PC12) cell apoptosis by the MTT assay and Hoechst staining. A serine/threonine kinase inhibitor attenuated this protection. G. elata resulted in phosphorylation and dephosphorylation of ERK1/2 and JNK1/2-p38 MAPKs (members of the serine/threonine kinase family), respectively, as revealed by Western blot analysis. An upstream ERK inhibitor attenuated G. elata-induced ERK phosphorylation but not protective effect. Although JNK and p38 inhibitors attenuated their related enzyme activities during serum deprivation, only JNK inhibitor prevented serum-deprived apoptosis. Thus, G. elata prevents serum-deprived apoptosis through activation of the serine/threonine kinase-dependent pathway and suppression of JNK activity.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Neuroprotective Agents/pharmacology , Analysis of Variance , Animals , Blotting, Western , Dicumarol/pharmacology , Flavonoids/pharmacology , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells , Phosphorylation , Pyridines/pharmacology , Rats , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles
19.
Biofactors ; 10(2-3): 131-8, 1999.
Article in English | MEDLINE | ID: mdl-10609874

ABSTRACT

Ubiquinone (UQ) reductase responsible for reduction of non-mitochondrial UQ was investigated in rats toward demonstrating an antioxidant role of UQ. In the liver, most of cellular UQ-10 reductase activity was attributable to a novel NADPH-UQ reductase in cytosol. The enzyme was not inhibited by dicumarol and rotenone, and had a Km of 19 microM for NADPH and 307 microM for NADH at the optimum pH 7.4. The enzyme was purified 300-fold to apparent homogeneity from the liver cytosol by an affinity chromatographic method. The purified enzyme reduced UQ-10 in lecithin liposomes, and protected the liposomes from lipid peroxidation. Furthermore, supplementation of rats with UQ-10 was observed to increase the enzyme level in their livers without affecting levels of other antioxidant factors. The observations suggested that cytosolic NADPH-UQ reductase is responsible for cellular UQ redox cycle as an endogenous antioxidant.


Subject(s)
Antioxidants/metabolism , Liver/enzymology , NADH, NADPH Oxidoreductases/metabolism , Ubiquinone/metabolism , Animals , Antimycin A/pharmacology , Antioxidants/chemistry , Cytosol/enzymology , Dicumarol/pharmacology , Electron Transport Complex I , Kinetics , Male , Models, Chemical , NAD/metabolism , NADP/metabolism , Organelles/enzymology , Oxidation-Reduction , Rats , Rats, Wistar , Rotenone/pharmacology , Ubiquinone/chemistry
20.
J Am Vet Med Assoc ; 212(6): 857-9, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9530428

ABSTRACT

Eight of 600 Holstein heifers and cows died after ingestion of sweet clover silage (Melilotus sp) that contained excessive concentrations of dicumarol caused by mold infestation. The cattle developed subcutaneous hemorrhages and bled from the vagina, became weak, were unable to move, and died. To the best of our knowledge, this is the first report of sweet clover poisoning in cattle from California and is discussed in light of previous findings in the Midwest and Canada. Sweet clover poisoning is caused by dicumarol, a fungal metabolite produced from substrates in sweet clover, and is a common livestock problem in the Northern Plains and Canada. Sweet clover poisoning should be considered in livestock animals with clinical evidence of hemostatic dysfunction, prolonged coagulation times, subcutaneous hemorrhages, and hemorrhagic abortions. Definite diagnosis of moldy sweet clover poisoning can be accomplished by analysis of serum and feed samples for dicumarol concentrations.


Subject(s)
Anticoagulants/analysis , Cattle Diseases/etiology , Dicumarol/analysis , Fabaceae/poisoning , Plant Poisoning/veterinary , Plants, Medicinal , Silage/poisoning , Animals , Anticoagulants/poisoning , California , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/mortality , Dicumarol/poisoning , Fabaceae/microbiology , Female , Hemorrhage/etiology , Hemorrhage/veterinary , Plant Poisoning/complications , Plant Poisoning/diagnosis , Silage/analysis , Silage/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL