Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Zhongguo Zhen Jiu ; 41(6): 683-4, 2021 Jun 12.
Article in Chinese | MEDLINE | ID: mdl-34085489

ABSTRACT

On the base of the principle of penetrating moxibustion and in combination with free adjustment devices such as movable U-shaped moxa stick holder and movable clamp, a new type of moxibustion box exerted on the head is designed, with precise positioning and sufficient heat intensity. Baihui moxibustion box is composed of two sections, i.e. body section and pillow section, which is as one structure. There are several vertical bar-shaped holes distributed evenly on the movable door outside moxa box. The U-shaped moxa stick holder on the inner side of the bar-shaped hole is connected with the fixed clamp on the outside, which is movable up and down, forward and backward for height adjustment. Such moxibustion box is characterized as accurate positioning, energy saving, temperature control and manpower saving.


Subject(s)
Moxibustion , Hot Temperature , Temperature , Workforce
2.
Shanghai Kou Qiang Yi Xue ; 30(2): 187-190, 2021 Apr.
Article in Chinese | MEDLINE | ID: mdl-34109360

ABSTRACT

PURPOSE: To compare apical root resorption of maxillary incisors between adolescents and adults after orthodontic treatment. METHODS: Patients receiving orthodontic treatment in Affiliated Hospital of Chifeng University from May 2014 to August 2016 were enrolled, and divided into two age groups: adolescent group (32) and adult group (36). The included subjects received orthodontic fixed appliance treatment with straight-wire technique combined with Hawley type retainer for one year. After treatment, all patients were followed up for one year. Then the apical root resorption of maxillary incisors was evaluated by cone-beam CT (CBCT) at 4 time points, including pre-treatment (T1), end of treatment (T2), 6 months after treatment (T3), and 12 months after treatment (T4). Data were processed by SPSS 20.0 software package. RESULTS: The external root volume of maxillary central incisor, lateral incisors, mandibular central incisors and mandibular lateral incisors of both sides at T2-T4 was significantly lower than that at T1(P<0.05). There was partial increase in root volume of both groups at T3 and T4, while no significant difference from that at T2 (P>0.05). △root volume T3-T2 and △root volume T4-T3 had no significant difference between the two groups (P>0.05). △root volume T3-T2 in the adolescent group was significantly smaller than that in the adult group (P<0.05). Correlation analysis showed that the △root volumeT1-T2 was significantly positively correlated with age (P<0.05), meanwhile △root volume T3-T2 and △root volume T4-T3 were negatively correlated with age (P<0.05). CONCLUSIONS: Age is an important factor affecting the volume of root after orthodontic treatment. Adolescent patients with Class II division 1 malocclusion have a strong ability of self-healing after orthodontic treatment.


Subject(s)
Malocclusion, Angle Class II , Root Resorption , Adolescent , Adult , Cone-Beam Computed Tomography , Humans , Incisor/diagnostic imaging , Maxilla/diagnostic imaging , Root Resorption/diagnostic imaging , Tooth Apex
3.
Acta Pharm Sin B ; 6(5): 492-503, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27709018

ABSTRACT

We have shown recently that concurrent harmaline, a monoamine oxidase-A inhibitor (MAOI), potentiates serotonin (5-HT) receptor agonist 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT)-induced hyperthermia. The objective of this study was to develop an integrated pharmacokinetic/pharmacodynamic (PK/PD) model to characterize and predict the thermoregulatory effects of such serotonergic drugs in mice. Physiological thermoregulation was described by a mechanism-based indirect-response model with adaptive feedback control. Harmaline-induced hypothermia and 5-MeO-DMT-elicited hyperthermia were attributable to the loss of heat through the activation of 5-HT1A receptor and thermogenesis via the stimulation of 5-HT2A receptor, respectively. Thus serotonergic 5-MeO-DMT-induced hyperthermia was readily distinguished from handling/injection stress-provoked hyperthermic effects. This PK/PD model was able to simultaneously describe all experimental data including the impact of drug-metabolizing enzyme status on 5-MeO-DMT and harmaline PK properties, and drug- and stress-induced simple hypo/hyperthermic and complex biphasic effects. Furthermore, the modeling results revealed a 4-fold decrease of apparent SC50 value (1.88-0.496 µmol/L) for 5-MeO-DMT when harmaline was co-administered, providing a quantitative assessment for the impact of concurrent MAOI harmaline on 5-MeO-DMT-induced hyperthermia. In addition, the hyperpyrexia caused by toxic dose combinations of harmaline and 5-MeO-DMT were linked to the increased systemic exposure to harmaline rather than 5-MeO-DMT, although the body temperature profiles were mispredicted by the model. The results indicate that current PK/PD model may be used as a new conceptual framework to define the impact of serotonergic agents and stress factors on thermoregulation.

4.
Pharmacol Rep ; 68(3): 608-15, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26977821

ABSTRACT

BACKGROUND: 5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and harmaline are indolealkylamine (IAA) drugs often abused together. Our recent studies have revealed the significant effects of co-administered harmaline, a monoamine oxidase inhibitor (MAOI), on 5-MeO-DMT pharmacokinetics and thermoregulation. This study was to delineate the impact of harmaline and 5-MeO-DMT on home-cage activity in mouse models, as well as the contribution of serotonin (5-HT) receptors. METHODS: Home-cage activities of individual animals were monitored automatically in the home cages following implantation of telemetry transmitters and administration of various doses of IAA drugs and 5-HT receptor antagonists. Area under the effect curve (AUEC) of mouse activity values were calculated by trapezoidal rule. RESULTS: High dose of harmaline (15mg/kg, ip) alone caused an early-phase (0-45min) hypoactivity in mice that was fully attenuated by 5-HT1A receptor antagonist WAY-100635, whereas a late-phase (45-180min) hyperactivity that was reduced by 5-HT2A receptor antagonist MDL-100907. 5-MeO-DMT (10 and 20mg/kg, ip) alone induced biphasic effects, an early-phase (0-45min) hypoactivity that was completely attenuated by WAY-100635, and a late-phase (45-180min) hyperactivity that was fully suppressed by MDL-100907. Interestingly, co-administration of MAOI harmaline (2-15mg/kg) with a subthreshold dose of 5-MeO-DMT (2mg/kg) induced excessive hyperactivities at late phase (45-180min) that could be abolished by either WAY-100635 or MDL-100907. CONCLUSIONS: Co-administration of MAOI with 5-MeO-DMT provokes excessive late-phase hyperactivity, which involves the activation of both 5-HT1A and 5-HT2A receptors.


Subject(s)
Harmaline/pharmacology , Hyperkinesis/chemically induced , Hypokinesia/chemically induced , Methoxydimethyltryptamines/pharmacology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Synergism , Fluorobenzenes , Harmaline/antagonists & inhibitors , Male , Methoxydimethyltryptamines/antagonists & inhibitors , Mice , Monoamine Oxidase Inhibitors/pharmacology , Motor Activity/drug effects , Piperazines/pharmacology , Piperidines , Pyridines/pharmacology
5.
Eur J Pharm Sci ; 82: 64-78, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26524713

ABSTRACT

Clopidogrel (Plavix®), is a widely used antiplatelet agent, which shows high inter-individual variability in treatment response in patients following the standard dosing regimen. In this study, a physiology-directed population pharmacokinetic/pharmacodynamic (PK/PD) model was developed based on clopidogrel and clopidogrel active metabolite (clop-AM) data from the PAPI and the PGXB2B studies using a step-wise approach in NONMEM (version 7.2). The developed model characterized the in vivo disposition of clopidogrel, its bioactivation into clop-AM in the liver and subsequent platelet aggregation inhibition in the systemic circulation reasonably well. It further allowed the identification of covariates that significantly impact clopidogrel's dose-concentration-response relationship. In particular, CYP2C19 intermediate and poor metabolizers converted 26.2% and 39.5% less clopidogrel to clop-AM, respectively, compared to extensive metabolizers. In addition, CES1 G143E mutation carriers have a reduced CES1 activity (82.9%) compared to wild-type subjects, which results in a significant increase in clop-AM formation. An increase in BMI was found to significantly decrease clopidogrel's bioactivation, whereas increased age was associated with increased platelet reactivity. Our PK/PD model analysis suggests that, in order to optimize clopidogrel dosing on a patient-by-patient basis, all of these factors have to be considered simultaneously, e.g. by using quantitative clinical pharmacology tools.


Subject(s)
Models, Biological , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/pharmacokinetics , Ticlopidine/analogs & derivatives , Adult , Clopidogrel , Demography , Female , Genotype , Humans , Liver/metabolism , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Ticlopidine/blood , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
6.
Clin Pharmacokinet ; 54(2): 147-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559342

ABSTRACT

Acute coronary syndromes (ACS) remain life-threatening disorders, which are associated with high morbidity and mortality. Dual antiplatelet therapy with aspirin and clopidogrel has been shown to reduce cardiovascular events in patients with ACS. However, there is substantial inter-individual variability in the response to clopidogrel treatment, in addition to prolonged recovery of platelet reactivity as a result of irreversible binding to P2Y12 receptors. This high inter-individual variability in treatment response has primarily been associated with genetic polymorphisms in the genes encoding for cytochrome (CYP) 2C19, which affect the pharmacokinetics of clopidogrel. While the US Food and Drug Administration has issued a boxed warning for CYP2C19 poor metabolizers because of potentially reduced efficacy in these patients, results from multivariate analyses suggest that additional factors, including age, sex, obesity, concurrent diseases and drug-drug interactions, may all contribute to the overall between-subject variability in treatment response. However, the extent to which each of these factors contributes to the overall variability, and how they are interrelated, is currently unclear. The objective of this review article is to provide a comprehensive update on the different factors that influence the pharmacokinetics and pharmacodynamics of clopidogrel and how they mechanistically contribute to inter-individual differences in the response to clopidogrel treatment.


Subject(s)
Ticlopidine/analogs & derivatives , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/metabolism , Clopidogrel , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Dose-Response Relationship, Drug , Humans , Polymorphism, Genetic , Precision Medicine , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacology , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
7.
Neuropharmacology ; 89: 342-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25446678

ABSTRACT

5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and harmaline are serotonin (5-HT) analogs often abused together, which alters thermoregulation that may indicate the severity of serotonin toxicity. Our recent studies have revealed that co-administration of monoamine oxidase inhibitor harmaline leads to greater and prolonged exposure to 5-HT agonist 5-MeO-DMT that might be influenced by cytochrome P450 2D6 (CYP2D6) status. This study was to define the effects of harmaline and 5-MeO-DMT on thermoregulation in wild-type and CYP2D6-humanized (Tg-CYP2D6) mice, as well as the involvement of 5-HT receptors. Animal core body temperatures were monitored noninvasively in the home cages after implantation of telemetry transmitters and administration of drugs. Harmaline (5 and 15 mg/kg, i.p.) alone was shown to induce hypothermia that was significantly affected by CYP2D6 status. In contrast, higher doses of 5-MeO-DMT (10 and 20 mg/kg) alone caused hyperthermia. Co-administration of harmaline (2, 5 or 15 mg/kg) remarkably potentiated the hyperthermia elicited by 5-MeO-DMT (2 or 10 mg/kg), which might be influenced by CYP2D6 status at certain dose combination. Interestingly, harmaline-induced hypothermia was only attenuated by 5-HT1A receptor antagonist WAY-100635, whereas 5-MeO-DMT- and harmaline-5-MeO-DMT-induced hyperthermia could be suppressed by either WAY-100635 or 5-HT2A receptor antagonists (MDL-100907 and ketanserin). Moreover, stress-induced hyperthermia under home cage conditions was not affected by WAY-100635 but surprisingly attenuated by MDL-100907 and ketanserin. Our results indicate that co-administration of monoamine oxidase inhibitor largely potentiates 5-MeO-DMT-induced hyperthermia that involves the activation of both 5-HT1A and 5-HT2A receptors. These findings shall provide insights into development of anxiolytic drugs and new strategies to relieve the lethal hyperthermia in serotonin toxicity.


Subject(s)
Fever/chemically induced , Harmaline/pharmacology , Methoxydimethyltryptamines/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Body Weight/drug effects , Cytochrome P-450 CYP2D6/genetics , Dose-Response Relationship, Drug , Drug Synergism , Fever/genetics , Fever/metabolism , Male , Mice , Mice, Transgenic , Serotonin Agents/pharmacology
8.
Drug Metab Dispos ; 41(5): 975-86, 2013 May.
Article in English | MEDLINE | ID: mdl-23393220

ABSTRACT

5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT or street name "5-MEO") is a newer designer drug belonging to a group of naturally occurring indolealkylamines. Our recent study has demonstrated that coadministration of monoamine oxidase A (MAO-A) inhibitor harmaline (5 mg/kg) increases systemic exposure to 5-MeO-DMT (2 mg/kg) and active metabolite bufotenine. This study is aimed at delineating harmaline and 5-MeO-DMT pharmacokinetic (PK) interactions at multiple dose levels, as well as the impact of CYP2D6 that affects harmaline PK and determines 5-MeO-DMT O-demethylation to produce bufotenine. Our data revealed that inhibition of MAO-A-mediated metabolic elimination by harmaline (2, 5, and 15 mg/kg) led to a sharp increase in systemic and cerebral exposure to 5-MeO-DMT (2 and 10 mg/kg) at all dose combinations. A more pronounced effect on 5-MeO-DMT PK was associated with greater exposure to harmaline in wild-type mice than CYP2D6-humanized (Tg-CYP2D6) mice. Harmaline (5 mg/kg) also increased blood and brain bufotenine concentrations that were generally higher in Tg-CYP2D6 mice. Surprisingly, greater harmaline dose (15 mg/kg) reduced bufotenine levels. The in vivo inhibitory effect of harmaline on CYP2D6-catalyzed bufotenine formation was confirmed by in vitro study using purified CYP2D6. Given these findings, a unified PK model including the inhibition of MAO-A- and CYP2D6-catalyzed 5-MeO-DMT metabolism by harmaline was developed to describe blood harmaline, 5-MeO-DMT, and bufotenine PK profiles in both wild-type and Tg-CYP2D6 mouse models. This PK model may be further employed to predict harmaline and 5-MeO-DMT PK interactions at various doses, define the impact of CYP2D6 status, and drive harmaline-5-MeO-DMT pharmacodynamics.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Harmaline/pharmacokinetics , Methoxydimethyltryptamines/pharmacokinetics , Monoamine Oxidase Inhibitors/pharmacokinetics , Animals , Catalysis , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2D6/genetics , Dose-Response Relationship, Drug , Harmaline/blood , Mice , Mice, Transgenic , Monoamine Oxidase/drug effects , Monoamine Oxidase Inhibitors/blood , Tandem Mass Spectrometry
9.
Curr Drug Metab ; 12(10): 997-1006, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22023319

ABSTRACT

Extrapolation of the metabolic, pharmacokinetic and toxicological data obtained from animals to humans is not always straightforward, given the remarkable species difference in drug metabolism that is due in large part to the differences in drug-metabolizing enzymes between animals and humans. Furthermore, genetic variations in drug-metabolizing enzymes may significantly alter pharmacokinetics, drug efficacy and safety. Thus, humanized transgenic mouse lines, in which the human drug-metabolizing enzymes are expressed in mouse tissues in the presence or absence of mouse orthologues, have been developed to address such challenges. These humanized transgenic mice are valuable animal models in understanding the significance of specific human drug-metabolizing enzymes in drug clearance and pharmacokinetics, as well as in predicting potential drug-drug interactions and chemical toxicity in humans. This review, therefore, aims to summarize the development and application of some humanized transgenic mouse models expressing human drug-metabolizing enzymes. The limitations of these genetically modified mouse models are also discussed.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Cytochrome P-450 Enzyme System/genetics , Glucuronosyltransferase/genetics , Mice, Transgenic , Pharmaceutical Preparations/metabolism , Animals , Arylamine N-Acetyltransferase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Drug Design , Glucuronosyltransferase/metabolism , Humans , Mice , Models, Animal , Pharmacokinetics
10.
Drug Metab Dispos ; 39(7): 1227-34, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21464174

ABSTRACT

5-Methoxy-N,N,-dimethyltryptamine (5-MeO-DMT), an abused serotonergic indolealkylamine drug, was placed into Schedule I controlled substance status in the United States as of January 19, 2011. In previous studies, we have shown the impact of monoamine oxidase A and cytochrome P450 2D6 enzymes on 5-MeO-DMT metabolism and pharmacokinetics. The aim of this study was to investigate 5-MeO-DMT pharmacokinetic properties after intravenous or intraperitoneal administration of three different doses (2, 10, and 20 mg/kg) to CYP2D6-humanized (Tg-CYP2D6) and wild-type control mice. Systemic exposure [area under the curve (AUC)] to 5-MeO-DMT was increased nonproportionally with the increase in dose. The existence of nonlinearity in serum 5-MeO-DMT pharmacokinetics was clearly manifested by dose-normalized AUC values, which were approximately 1.5- to 2.0-fold (intravenous) and 1.8- to 2.7-fold (intraperitoneal) higher in wild-type or Tg-CYP2D6 mice dosed with 10 and 20 mg/kg 5-MeO-DMT, respectively, than those in mice treated with 2 mg/kg 5-MeO-DMT. Furthermore, a two-compartment model including first-order absorption, nonlinear (Michaelis-Menten) elimination, and CYP2D6-dependent linear elimination from the central compartment was developed to characterize the intravenous and intraperitoneal pharmacokinetic data for 5-MeO-DMT in wild-type and Tg-CYP2D6 mice. In addition, 5-MeO-DMT was readily detected in mouse brain after drug treatment, and brain 5-MeO-DMT concentrations were also increased nonproportionally with the increase of dose. The results establish a nonlinear pharmacokinetic property for 5-MeO-DMT in mice, suggesting that the risk of 5-MeO-DMT intoxication may be increased nonproportionally at higher doses.


Subject(s)
Methoxydimethyltryptamines/pharmacokinetics , Animals , Area Under Curve , Brain/metabolism , Chromatography, Liquid , Methoxydimethyltryptamines/blood , Mice , Tandem Mass Spectrometry
11.
Drug Metab Rev ; 43(1): 27-40, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20854191

ABSTRACT

Determining the in vivo significance of a specific enzyme, transporter, or xenobiotic receptor in drug metabolism and pharmacokinetics may be hampered by gene multiplicity and complexity, levels of expression, and interaction between various components involved. The development of knockout (loss-of-function) and transgenic (gain-of-function) mouse models opens the door to the improved understanding of gene function in a whole-body system. There is also growing interest in the development of humanized mice to overcome species differences in drug metabolism and disposition. This review, therefore, aims to summarize and discuss some successful examples of drug-metabolizing enzyme, transporter, and nuclear-receptor genetically modified mouse models. These genetically modified mouse models have been proven as invaluable models for understanding in vivo function of drug-metabolizing enzymes, transporters, and xenobiotic receptors in drug metabolism and transport, as well as predicting potential drug-drug interaction and toxicity in humans. Nevertheless, concerns remain about interpretation of data obtained from such genetically modified mouse models, in which the expression of related genes is altered significantly.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Membrane Transport Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Xenobiotics/pharmacokinetics , Animals , Animals, Genetically Modified , Cytochrome P-450 Enzyme System/genetics , Drug Discovery , Humans , Inactivation, Metabolic , Membrane Transport Proteins/genetics , Mice , Receptors, Cytoplasmic and Nuclear/genetics
12.
Curr Drug Metab ; 11(8): 659-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20942780

ABSTRACT

5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) belongs to a group of naturally-occurring psychoactive indolealkylamine drugs. It acts as a nonselective serotonin (5-HT) agonist and causes many physiological and behavioral changes. 5-MeO-DMT is O-demethylated by polymorphic cytochrome P450 2D6 (CYP2D6) to an active metabolite, bufotenine, while it is mainly inactivated through the deamination pathway mediated by monoamine oxidase A (MAO-A). 5-MeO-DMT is often used with MAO-A inhibitors such as harmaline. Concurrent use of harmaline reduces 5-MeO-DMT deamination metabolism and leads to a prolonged and increased exposure to the parent drug 5-MeO-DMT, as well as the active metabolite bufotenine. Harmaline, 5-MeO-DMT and bufotenine act agonistically on serotonergic systems and may result in hyperserotonergic effects or serotonin toxicity. Interestingly, CYP2D6 also has important contribution to harmaline metabolism, and CYP2D6 genetic polymorphism may cause considerable variability in the metabolism, pharmacokinetics and dynamics of harmaline and its interaction with 5-MeO-DMT. Therefore, this review summarizes recent findings on biotransformation, pharmacokinetics, and pharmacological actions of 5-MeO-DMT. In addition, the pharmacokinetic and pharmacodynamic drug-drug interactions between harmaline and 5-MeO-DMT, potential involvement of CYP2D6 pharmacogenetics, and risks of 5-MeO-DMT intoxication are discussed.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Hallucinogens/pharmacology , Methoxydimethyltryptamines/pharmacology , Animals , Bufotenin/metabolism , Bufotenin/pharmacology , Drug Interactions , Hallucinogens/pharmacokinetics , Hallucinogens/toxicity , Harmaline/pharmacology , Humans , Methoxydimethyltryptamines/pharmacokinetics , Methoxydimethyltryptamines/toxicity , Pharmacogenetics , Serotonin Receptor Agonists/pharmacokinetics , Serotonin Receptor Agonists/pharmacology , Serotonin Receptor Agonists/toxicity
13.
Biochem Pharmacol ; 80(1): 122-8, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20206139

ABSTRACT

5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is a natural psychoactive indolealkylamine drug that has been used for recreational purpose. Our previous study revealed that polymorphic cytochrome P450 2D6 (CYP2D6) catalyzed 5-MeO-DMT O-demethylation to produce active metabolite bufotenine, while 5-MeO-DMT is mainly inactivated through deamination pathway mediated by monoamine oxidase (MAO). This study, therefore, aimed to investigate the impact of CYP2D6 genotype/phenotype status and MAO inhibitor (MAOI) on 5-MeO-DMT metabolism and pharmacokinetics. Enzyme kinetic studies using recombinant CYP2D6 allelic isozymes showed that CYP2D6.2 and CYP2D6.10 exhibited 2.6- and 40-fold lower catalytic efficiency (V(max)/K(m)), respectively, in producing bufotenine from 5-MeO-DMT, compared with wild-type CYP2D6.1. When co-incubated with MAOI pargyline, 5-MeO-DMT O-demethylation in 10 human liver microsomes showed significantly strong correlation with bufuralol 1'-hydroxylase activities (R(2)=0.98; P<0.0001) and CYP2D6 contents (R(2)=0.77; P=0.0007), whereas no appreciable correlations with enzymatic activities of other P450 enzymes. Furthermore, concurrent MAOI harmaline sharply reduced 5-MeO-DMT depletion and increased bufotenine formation in human CYP2D6 extensive metabolizer hepatocytes. In vivo studies in wild-type and CYP2D6-humanized (Tg-CYP2D6) mouse models showed that Tg-CYP2D6 mice receiving the same dose of 5-MeO-DMT (20mg/kg, i.p.) had 60% higher systemic exposure to metabolite bufotenine. In addition, pretreatment of harmaline (5mg/kg, i.p.) led to 3.6- and 4.4-fold higher systemic exposure to 5-MeO-DMT (2mg/kg, i.p.), and 9.9- and 6.1-fold higher systemic exposure to bufotenine in Tg-CYP2D6 and wild-type mice, respectively. These findings indicate that MAOI largely affects 5-MeO-DMT metabolism and pharmacokinetics, as well as bufotenine formation that is mediated by CYP2D6.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Methoxydimethyltryptamines/metabolism , Methoxydimethyltryptamines/pharmacokinetics , Monoamine Oxidase Inhibitors/pharmacology , Psychotropic Drugs/metabolism , Animals , Area Under Curve , Bufotenin/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 Enzyme System/pharmacology , Dose-Response Relationship, Drug , Genotype , Half-Life , Harmaline/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Methoxydimethyltryptamines/pharmacology , Methylation/drug effects , Mice , Mice, Transgenic , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Monoamine Oxidase/metabolism , Pargyline/metabolism , Phenotype , Polymorphism, Genetic/drug effects , Psychotropic Drugs/pharmacology
14.
Biochem Pharmacol ; 78(6): 617-24, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19445902

ABSTRACT

Harmaline is a beta-carboline alkaloid showing neuroprotective and neurotoxic properties. Our recent studies have revealed an important role for cytochrome P450 2D6 (CYP2D6) in harmaline O-demethylation. This study, therefore, aimed to delineate the effects of CYP2D6 phenotype/genotype on harmaline metabolism, pharmacokinetics (PK) and pharmacodynamics (PD), and to develop a pharmacogenetics mechanism-based compartmental PK model. In vitro kinetic studies on metabolite formation in human CYP2D6 extensive metabolizer (EM) and poor metabolizer (PM) hepatocytes indicated that harmaline O-demethylase activity (V(max)/K(m)) was about 9-fold higher in EM hepatocytes. Substrate depletion showed mono-exponential decay trait, and estimated in vitro harmaline clearance (CL(int), microL/min/10(6)cells) was significantly lower in PM hepatocytes (28.5) than EM hepatocytes (71.1). In vivo studies in CYP2D6-humanized and wild-type mouse models showed that wild-type mice were subjected to higher and longer exposure to harmaline (5 and 15mg/kg; i.v. and i.p.), and more severe hypothermic responses. The PK/PD data were nicely described by our pharmacogenetics-based PK model involving the clearance of drug by CYP2D6 (CL(CYP2D6)) and other mechanisms (CL(other)), and an indirect response PD model, respectively. Wild-type mice were also more sensitive to harmaline in marble-burying tests, as manifested by significantly lower ED(50) and steeper Hill slope. These findings suggest that distinct CYP2D6 status may cause considerable variations in harmaline metabolism, PK and PD. In addition, the pharmacogenetics-based PK model may be extended to define PK difference caused by other polymorphic drug-metabolizing enzyme in different populations.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Harmaline/metabolism , Microsomes, Liver/metabolism , Animals , Behavior/drug effects , Behavior/physiology , Genotype , Harmaline/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Transgenic , Microsomes, Liver/drug effects , Pharmacogenetics , Phenotype , Polymorphism, Genetic
15.
Drug Metab Dispos ; 37(3): 443-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19095720

ABSTRACT

Pinoline, 6-methoxy-1,2,3,4-tetrahydro-beta-carboline, is a serotonin analog that selectively inhibits the activity of monoamine oxidase-A and shows antidepressant activity. Our previous study using a panel of recombinant cytochrome P450 (P450) enzymes suggests that pinoline O-demethylation may be selectively catalyzed by polymorphic CYP2D6. The current study, therefore, aimed to delineate the impact of CYP2D6 status on pinoline metabolism. Enzyme kinetic studies using recombinant CYP2D6 allelic isozymes revealed that CYP2D6.2 exhibited 5-fold lower enzyme efficiency (V(max)/K(m)) toward pinoline compared with CYP2D6.1, and CYP2D6.10 did not show any catalytic activity. Inhibition study showed that quinidine (1 microM) completely blocked pinoline O-demethylase activity in human liver microsomes, whereas other P450 isoform-selective inhibitors had no or minimal effects. Pinoline O-demethylase activities in 10 human liver microsomes showed significantly strong correlation with bufuralol 1'-hydroxylase activities (R(2)=0.93; p<0.0001) and CYP2D6 contents (R(2)=0.82; p=0.005), whereas no appreciable correlations with enzymatic activities of other P450 enzymes were found. Furthermore, we compared pinoline urinary metabolic ratio (pinoline/6-hydroxy-1,2,3,4-tetrahydro-beta-carboline) between CYP2D6-humanized and wild-type control mice after intraperitoneal injection of pinoline (30 mg/kg). Results indicated that the two genotyped mice were clearly distinguished by pinoline metabolic ratio (mean +/- S.D.), which was much higher in wild-type mice (0.29+/-0.19, n=4) than in CYP2D6-humanized transgenic mice (0.0070+/-0.0048, n=4). Our findings suggest that pinoline O-demethylation is governed by CYP2D6 status, and pinoline, at a proper concentration or dose, may be a good probe to evaluate CYP2D6 activity.


Subject(s)
Carbolines/metabolism , Cytochrome P-450 CYP2D6/metabolism , Molecular Probes , Animals , Humans , Mice , Mice, Transgenic , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism
16.
Drug Metab Dispos ; 37(1): 170-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18832475

ABSTRACT

Accurate quantification of cytochrome P450 (P450) protein contents is essential for reliable assessment of drug safety, including the prediction of in vivo clearance from in vitro metabolism data, which may be hampered by the use of uncharacterized standards and existence of unknown allelic isozymes. Therefore, this study aimed to delineate the variability in absolute quantification of polymorphic CYP2D6 drug-metabolizing enzyme and compare immunoblot and nano liquid chromatography coupled to mass spectrometry (nano-LC/MS) methods in identification and relative quantification of CYP2D6.1 and CYP2D6.2 allelic isozymes. Holoprotein content of in-house purified CYP2D6 isozymes was determined according to carbon monoxide difference spectrum, and total protein was quantified with bicinchoninic acid protein assay. Holoprotein/total CYP2D6 protein ratio was markedly higher for purified CYP2D6.1 (71.0%) than that calculated for CYP2D6.1 Supersomes (35.5%), resulting in distinct linear calibration range (0.05-0.50 versus 0.025-0.25 pmol) that was determined by densitometric analysis of immunoblot bands. Likewise, purified CYP2D6.2 and CYP2D6.10 and the CYP2D6.10 Supersomes all showed different holoprotein/total CYP2D6 protein ratios and distinct immunoblot linear calibration ranges. In contrast to immunoblot, nano-LC/MS readily distinguished CYP2D6.2 (R296C and S486T) from CYP2D6.1 by isoform-specific proteolytic peptides that contain the altered amino acid residues. In addition, relative quantitation of the two allelic isozymes was successfully achieved with label-free protein quantification, consistent with the nominated ratio. Because immunoblot and nano-LC/MS analyses measure total P450 protein (holoprotein and apoprotein) in a sample, complete understanding of holoprotein and apoprotein contents in P450 standards is desired toward reliable quantification. Our data also suggest that nano-LC/MS not only facilitates P450 quantitation but also provides genotypic information.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Isoenzymes/metabolism , Alleles , Amino Acid Sequence , Blotting, Western , Chromatography, Liquid , Cytochrome P-450 CYP2D6/chemistry , Cytochrome P-450 CYP2D6/genetics , Electrophoresis, Polyacrylamide Gel , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Mass Spectrometry , Molecular Sequence Data
17.
Bioanalysis ; 1(1): 87-95, 2009 Apr.
Article in English | MEDLINE | ID: mdl-20523750

ABSTRACT

INTRODUCTION: 5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is a psychoactive indolealkylamine substance that has been used for recreational purpose and may lead to fatal toxicity. While 5-MeO-DMT is mainly inactivated via deamination, it is O-demethylated to an active metabolite, bufotenine. Quantitation of 5-MeO-DMT and bufotenine is essential to understand the exposure to and the effects of drug and metabolite. This study, therefore, aimed to develop and validate a LC-MS/MS method for simultaneous analysis of 5-MeO-DMT and bufotenine in mouse serum. METHODS: A simple protein precipitation method coupled with an optimal gradient elution was used for sample preparation and separation. Detection of 5-MeO-DMT and bufotenine was accomplished using multiple reaction monitoring of m/z 219.2→174.2 and 205.2→160.2, respectively, in the positive ion mode. 5-Methyl-N,N-dimethyltrypamine (m/z 203.2→158.3) was used as internal standard for quantification. Accuracy and precision were determined after the analyses of quality control samples. Validated assay was then employed to determine drug and metabolite concentrations in serum samples collected from mice at different time points after intraperitoneal administration of 5-MeO-DMT (2 mg/kg). RESULTS: With a total run time of 9 min, 5-MeO-DMT and bufotenine were eluted at 2.8 and 5.6 min, respectively. The assay was linear over the range 0.90-5,890 ng/mL (1.12-7,360 pg on-column) for 5-MeO-DMT and 2.52-5,510 ng/mL (3.14-6,890 pg) for bufotenine. Intra- and inter-day precision and accuracy were within 15% for both analytes. The recovery of each analyte from 20 µL of serum containing 8.08, 72.7 and 655 ng/mL of 5-MeO-DMT and 7.56, 68.1 and 613 ng/mL of bufotenine was more than 75%. Pharmacokinetic analysis revealed that the systemic exposure (area under the curve) to metabolite bufotenine was about 1/14 of that to 5-MeO-DMT. CONCLUSION: This LC-MS/MS method is a sensitive and reliable assay for quantitation of blood 5-MeO-DMT and bufotenine. Given the fact that bufotenine acts on 5-HT(2A) receptor with an affinity about 10-fold higher than 5-MeO-DMT, the active metabolite bufotenine may significantly contribute to the apparent pharmacological and toxicological effects of 5-MeO-DMT.

18.
Article in English | MEDLINE | ID: mdl-17267298

ABSTRACT

A sensitive liquid chromatography-mass spectrometric (LC/MS) method for the quantification of ginsenoside Rd in dog plasma was developed and validated after solid-phase extraction (SPE). Chromatographic separation was achieved on a reversed-phase Cromosil C(18) column with the mobile phase of acetonitrile-ammonium chloride (500 micromol/L) and step gradient elution resulted in a total run time of about 5.5 min. The analytes were detected by using an electrospray negative ionization mass spectrometry in the selected ion monitoring (SIM) mode. A good linear relationship was obtained in the concentration range studied (0.005-2.500 microg/mL) (r=0.9998). Lower limit of quantification (LLOQ) was 5 ng/mL by using 500 microL plasma sample. Average recoveries ranged from 70.71 to 75.89% in plasma at the concentrations of 0.010, 0.100 and 2.500 microg/mL. Intra- and inter-day relative standard deviations were 8.49-11.71 and 5.71-16.48%, respectively. This method was successfully applied to the pharmacokinetic studies on dogs. The absolute bioavailability of Rd in dogs was 0.26%.


Subject(s)
Chromatography, Liquid/methods , Ginsenosides/blood , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Biological Availability , Dogs , Ginsenosides/pharmacokinetics , Sensitivity and Specificity
19.
Eur J Drug Metab Pharmacokinet ; 30(1-2): 63-7, 2005.
Article in English | MEDLINE | ID: mdl-16010863

ABSTRACT

To develop a HPLC-MS method of determining ginsenoside Rh2 in dog plasma based on solid-phase extraction for pharmacokinetic studies. Six dogs were randomly assigned to two groups, either given 0.1 mg/kg dose intravenously or 1 mg/kg dose through oral gavage. Analysis using high performance liquid chromatography (HPLC) with ODS column, followed by detection with electrospray ionization(ESI) mass spectrometry(MS) in negative ion mode with 500 microM ammonium chloride in the mobile phase. The assays were validated over the concentration range of 2.0-1250.0 ng/ml in dog plasma. The intra- and inter- day precision were less than 10% in terms of RSD. The overall recovery was more than 80%. The validated assay was suitable for pharmacokinetic studies of ginsenoside Rh2 and the observed oral bioavailabilities of Rh2 were 17.6% and 24.8% for male and female dogs respectively.


Subject(s)
Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/pharmacokinetics , Ginsenosides/analysis , Ginsenosides/pharmacokinetics , Animals , Chromatography, High Pressure Liquid , Dogs , Mass Spectrometry , Sensitivity and Specificity
20.
Article in English | MEDLINE | ID: mdl-15734156

ABSTRACT

To support pharmacokinetic studies of ginsenosides, a novel method to quantitatively analyze ginsenoside Rg3 (Rg3), its prosapogenin ginsenoside Rh2 (Rh2) and aglycone 20(S)-protopanaxadiol (ppd) in rat plasma was developed and validated. The method was based on gradient separation of ginsenosides present in rat plasma using high performance liquid chromatography (HPLC), followed by detection with electrospray ionization(ESI) mass spectrometry (MS) in negative ion mode with the mobile phase additive, ammonium chloride (500 microM). Differentiation of ginsenosides was achieved through simultaneous detection of the [M(+)Cl(-)] adduct of ginsenoside Rg3 and [M(+)Cl(-)] adducts of its deglycosylated metabolites Rh2 and ppd, and other ions after solid phase extraction (SPE). The /specific ions monitored were m/z 819.50 for Rg3, m/z 657.35 for Rh2, m/z 495.40 for ppd and m/z 799.55 for the internal standard (digitoxin). The mean recoveries for Rg3, Rh2 and ppd were 77.85, 82.65 and 98.33%, respectively using 0.1 ml plasma for extraction. The lower limits of quantification were 10.0, 2.0 and 8.0 ng/ml (equivalent to 0.1, 0.02 and 0.08 ng in each 10 microl injection onto the HPLC column) for Rg3, Rh2 and ppd, respectively. The method has been demonstrated to be highly sensitive and accurate for the determination of Rg3 and its metabolites in rat plasma.


Subject(s)
Chromatography, High Pressure Liquid/methods , Ginsenosides/blood , Ginsenosides/pharmacokinetics , Animals , Drug Stability , Freezing , Ginsenosides/isolation & purification , Rats , Rats, Sprague-Dawley , Sapogenins/blood , Sapogenins/isolation & purification , Spectrometry, Mass, Electrospray Ionization , Triterpenes/blood , Triterpenes/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...