Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Vet Pharmacol Ther ; 43(6): 608-613, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32893906

RESUMO

In humans, the cytochrome P450 3A (CYP3A) subfamily is involved in midazolam (MDZ) biotransformation into 1'- and 4-hydroxy metabolites, and the former serves as a probe for CYP3A catalytic activity. In veterinary species is still crucial to identify enzyme- and species-specific CYP substrates; thus, the aim of this study was to characterize MDZ oxidation in cattle liver. A HPLC-UV method was used to measure 1'- and 4-hydroxy MDZ (1'- and 4-OHMDZ, respectively) formation in cattle liver microsomes and assess the role of CYP3A by an immunoinhibition study. Moreover, MDZ hydroxylation was evaluated in 300 cattle liver samples and results were correlated with testosterone hydroxylation. Formation of both metabolites conformed to a single-enzyme Michaelis-Menten kinetics. Values of Vmax and Km were 0.67 nmol/min/mg protein and 6.16 µM for 4-OHMDZ, and 0.06 nmol/min/mg protein and 10.08 µM for 1'-OHMDZ. An anti-rat CYP3A1 polyclonal antibody inhibited up to 50% and 94% 1'- and 4-OHMDZ formation, respectively. MDZ oxidation in liver microsomes was poorly correlated with testosterone hydroxylation. In conclusion, cattle metabolized MDZ to 1'-OHMDZ and 4-OHMDZ. The immunoinhibition results indicated a major contribution of CYP3As to 4-OHMDZ formation and the involvement of other CYPs in 1'-OHMDZ production, paving the way for further investigations.


Assuntos
Adjuvantes Anestésicos/metabolismo , Bovinos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Microssomos Hepáticos/metabolismo , Midazolam/metabolismo , Animais , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Oxirredução
2.
Vet Anaesth Analg ; 47(2): 200-209, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31983556

RESUMO

OBJECTIVE: To estimate the pharmacokinetics of midazolam and 1-hydroxymidazolam after midazolam administration as an intravenous bolus in sevoflurane-anesthetized cats. STUDY DESIGN: Prospective pharmacokinetic study. ANIMALS: A group of six healthy adult, female domestic cats. METHODS: Anesthesia was induced and maintained with sevoflurane. After 30 minutes of anesthetic equilibration, cats were administered midazolam (0.3 mg kg-1) over 15 seconds. Venous blood was collected at 0, 1, 2, 4, 8, 15, 30, 45, 90, 180 and 360 minutes after administration. Plasma concentrations for midazolam and 1-hydroxymidazolam were measured using high-pressure liquid chromatography. The heart rate (HR), respiratory rate (fR), rectal temperature, noninvasive mean arterial pressure (MAP) and end-tidal carbon dioxide (Pe'CO2) were recorded at 5 minute intervals. Population compartment models were fitted to the time-plasma midazolam and 1-hydroxymidazolam concentrations using nonlinear mixed effect modeling. RESULTS: The pharmacokinetic model was fitted to the data from five cats, as 1-hydroxymidazolam was not detected in one cat. A five-compartment model best fitted the data. Typical values (% interindividual variability where estimated) for the volumes of distribution for midazolam (three compartments) and hydroxymidazolam (two compartments) were 117 (14), 286 (10), 705 (14), 53 (36) and 334 mL kg-1, respectively. Midazolam clearance to 1-hydroxymidazolam, midazolam fast and slow intercompartmental clearances, 1-hydroxymidazolam clearance and 1-hydroxymidazolam intercompartment clearance were 18.3, 63.5 (15), 22.1 (8), 1.7 (67) and 3.8 mL minute-1 kg-1, respectively. No significant changes in HR, MAP, fR or Pe'CO2 were observed following midazolam administration. CONCLUSION AND CLINICAL RELEVANCE: In sevoflurane-anesthetized cats, a five-compartment model best fitted the midazolam pharamacokinetic profile. There was a high interindividual variability in the plasma 1-hydroxymidazolam concentrations, and this metabolite had a low clearance and persisted in the plasma for longer than the parent drug. Midazolam administration did not result in clinically significant changes in physiologic variables.


Assuntos
Gatos/metabolismo , Midazolam/farmacocinética , Sevoflurano/farmacologia , Adjuvantes Anestésicos/administração & dosagem , Adjuvantes Anestésicos/metabolismo , Adjuvantes Anestésicos/farmacocinética , Anestésicos Inalatórios/administração & dosagem , Anestésicos Inalatórios/farmacologia , Animais , Gatos/fisiologia , Interações Medicamentosas , Feminino , Meia-Vida , Injeções Intravenosas/veterinária , Midazolam/administração & dosagem , Midazolam/metabolismo , Sevoflurano/administração & dosagem
3.
Pharm Res ; 35(8): 162, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29931580

RESUMO

PURPOSE: To investigate influence of inflammation on metabolism and pharmacokinetics (PK) of midazolam (MDZ) and construct a semi-physiologically based pharmacokinetic (PBPK) model to predict PK in mice with inflammatory disease. METHODS: Glucose-6-phosphate isomerase (GPI)-mediated inflammation was used as a preclinical model of arthritis in DBA/1 mice. CYP3A substrate MDZ was selected to study changes in metabolism and PK during the inflammation. The semi-PBPK model was constructed using mouse physiological parameters, liver microsome metabolism, and healthy animal PK data. In addition, serum cytokine, and liver-CYP (cytochrome P450 enzymes) mRNA levels were examined. RESULTS: The in vitro metabolite formation rate was suppressed in liver microsomes prepared from the GPI-treated mice as compared to the healthy mice. Further, clearance of MDZ was reduced during inflammation as compared to the healthy group. Finally, the semi-PBPK model was used to predict PK of MDZ after GPI-mediated inflammation. IL-6 and TNF-α levels were elevated and liver-cyp3a11 mRNA was reduced after GPI treatment. CONCLUSION: The semi-PBPK model successfully predicted PK parameters of MDZ in the disease state. The model may be applied to predict PK of other drugs under disease conditions using healthy animal PK and liver microsomal data as inputs.


Assuntos
Adjuvantes Anestésicos/farmacocinética , Inflamação/metabolismo , Midazolam/farmacocinética , Adjuvantes Anestésicos/metabolismo , Animais , Citocromo P-450 CYP3A/metabolismo , Glucose-6-Fosfato Isomerase/metabolismo , Humanos , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Midazolam/metabolismo , Modelos Biológicos
4.
J Pharm Sci ; 106(9): 2826-2838, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28495566

RESUMO

The use of in vitro-in vivo extrapolation (IVIVE) techniques, mechanistically incorporated within physiologically based pharmacokinetic (PBPK) models, can harness in vitro drug data and enhance understanding of in vivo pharmacokinetics. This study's objective was to develop a user-friendly rat (250 g, male Sprague-Dawley) IVIVE-linked PBPK model. A 13-compartment PBPK model including mechanistic absorption models was developed, with required system data (anatomical, physiological, and relevant IVIVE scaling factors) collated from literature and analyzed. Overall, 178 system parameter values for the model are provided. This study also highlights gaps in available system data required for strain-specific rat PBPK model development. The model's functionality and performance were assessed using previous literature-sourced in vitro properties for diazepam, metoprolol, and midazolam. The results of simulations were compared against observed pharmacokinetic rat data. Predicted and observed concentration profiles in 10 tissues for diazepam after a single intravenous (i.v.) dose making use of either observed i.v. clearance (CLiv) or in vitro hepatocyte intrinsic clearance (CLint) for simulations generally led to good predictions in various tissue compartments. Overall, all i.v. plasma concentration profiles were successfully predicted. However, there were challenges in predicting oral plasma concentration profiles for metoprolol and midazolam, and the potential reasons and according solutions are discussed.


Assuntos
Adjuvantes Anestésicos/farmacocinética , Antiarrítmicos/farmacocinética , Anticonvulsivantes/farmacocinética , Diazepam/farmacocinética , Metoprolol/farmacocinética , Midazolam/farmacocinética , Adjuvantes Anestésicos/administração & dosagem , Adjuvantes Anestésicos/sangue , Adjuvantes Anestésicos/metabolismo , Administração Intravenosa , Animais , Antiarrítmicos/administração & dosagem , Antiarrítmicos/sangue , Antiarrítmicos/metabolismo , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/sangue , Anticonvulsivantes/metabolismo , Simulação por Computador , Diazepam/administração & dosagem , Diazepam/sangue , Diazepam/metabolismo , Hepatócitos/metabolismo , Masculino , Taxa de Depuração Metabólica , Metoprolol/administração & dosagem , Metoprolol/sangue , Metoprolol/metabolismo , Midazolam/administração & dosagem , Midazolam/sangue , Midazolam/metabolismo , Modelos Biológicos , Ratos Sprague-Dawley
5.
Neurochem Int ; 109: 41-53, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28300620

RESUMO

GABA is catabolized in the mitochondrial matrix through the GABA shunt, encompassing transamination to succinic semialdehyde followed by oxidation to succinate by the concerted actions of GABA transaminase (GABA-T) and succinic semialdehyde dehydrogenase (SSADH), respectively. Gamma-hydroxybutyrate (GHB) is a neurotransmitter and a psychoactive drug that could enter the citric acid cycle through transhydrogenation with α-ketoglutarate to succinic semialdehyde and d-hydroxyglutarate, a reaction catalyzed by hydroxyacid-oxoacid transhydrogenase (HOT). Here, we tested the hypothesis that the elevation in matrix succinate concentration caused by exogenous addition of GABA, succinic semialdehyde or GHB shifts the equilibrium of the reversible reaction catalyzed by succinate-CoA ligase towards ATP (or GTP) hydrolysis, effectively negating substrate-level phosphorylation (SLP). Mitochondrial SLP was addressed by interrogating the directionality of the adenine nucleotide translocase during anoxia in isolated mouse brain and liver mitochondria. GABA eliminated SLP, and this was rescued by the GABA-T inhibitors vigabatrin and aminooxyacetic acid. Succinic semialdehyde was an extremely efficient substrate energizing mitochondria during normoxia but mimicked GABA in abolishing SLP in anoxia, in a manner refractory to vigabatrin and aminooxyacetic acid. GHB could moderately energize liver but not brain mitochondria consistent with the scarcity of HOT expression in the latter. In line with these results, GHB abolished SLP in liver but not brain mitochondria during anoxia and this was unaffected by either vigabatrin or aminooxyacetic acid. It is concluded that when mitochondria catabolize GABA or succinic semialdehyde or GHB through the GABA shunt, their ability to perform SLP is impaired.


Assuntos
Mitocôndrias/metabolismo , Oxibato de Sódio/metabolismo , Ácido gama-Aminobutírico/análogos & derivados , Ácido gama-Aminobutírico/metabolismo , Adjuvantes Anestésicos/metabolismo , Animais , Feminino , Masculino , Metabolismo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/fisiologia , Especificidade por Substrato/fisiologia
6.
J Pharm Sci ; 102(9): 3161-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23686764

RESUMO

Digoxin is the recommended substrate for assessment of P-glycoprotein (P-gp)-mediated drug-drug interactions (DDIs) in vivo. The overall aim of our study was to investigate the inhibitory potential of both verapamil and norverapamil on the P-gp-mediated efflux of digoxin in both gut and liver. Therefore, a physiologically-based pharmacokinetic (PBPK) model for verapamil and its primary metabolite was developed and validated through the recovery of observed clinical plasma concentration data for both moieties and the reported interaction with midazolam, albeit a cytochrome P450 3A4-mediated DDI. The validated inhibitor model was then used in conjunction with the model developed previously for digoxin. The range of values obtained for the 10 trials indicated that increases in area under the plasma concentration-time curve (AUC) profiles and maximum plasma concentration observed (Cmax ) values of digoxin following administration of verapamil were more comparable with in vivo observations, when P-gp inhibition by the metabolite, norverapamil, was considered as well. The predicted decrease in AUC and Cmax values of digoxin following administration of rifampicin because of P-gp induction was 1.57- (range: 1.42-1.77) and 1.62-fold (range: 1.53-1.70), which were reasonably consistent with observed values of 1.4- and 2.2-fold, respectively. This study demonstrates the application of permeability-limited models of absorption and distribution within a PBPK framework together with relevant in vitro data on transporters to assess the clinical impact of modulated P-gp-mediated efflux by drugs in development.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Antiarrítmicos/farmacologia , Antiarrítmicos/farmacocinética , Digoxina/farmacologia , Digoxina/farmacocinética , Verapamil/análogos & derivados , Verapamil/farmacologia , Adjuvantes Anestésicos/metabolismo , Adjuvantes Anestésicos/farmacologia , Antiarrítmicos/metabolismo , Simulação por Computador , Digoxina/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Midazolam/metabolismo , Midazolam/farmacologia , Modelos Biológicos , Permeabilidade/efeitos dos fármacos , Rifampina/metabolismo , Rifampina/farmacologia , Verapamil/metabolismo
7.
Chem Res Toxicol ; 26(3): 486-9, 2013 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-23419139

RESUMO

In vivo drug interactions of the teratogen thalidomide with the model cytochrome P450 (P450) 3A substrate midazolam were investigated in mice with humanized livers. The clearance of midazolam (administered intravenously, 10 mg kg(-1)) in chimeric mice was enhanced by orally co-administered thalidomide (100 mg kg(-1)). A larger area under the curve of the major metabolite 1'-hydroxymidazolam (1.7-fold) was obtained with thalidomide because of the heterotropic cooperativity of human P450 3A enzymes. A larger area under the curve of the minor metabolite 4-hydroxymidazolam (3.5-fold) was seen with daily pretreatment with thalidomide for 3 days, presumably because of human P450 3A induction. These results demonstrate that livers of humanized mice mediate drug interactions of thalidomide and suggest interactions of therapeutic agents during therapies with thalidomide.


Assuntos
Adjuvantes Anestésicos/farmacologia , Citocromo P-450 CYP3A/metabolismo , Imunossupressores/farmacologia , Fígado/metabolismo , Midazolam/farmacologia , Teratogênicos/farmacologia , Talidomida/farmacologia , Adjuvantes Anestésicos/administração & dosagem , Adjuvantes Anestésicos/sangue , Adjuvantes Anestésicos/metabolismo , Animais , Quimera/metabolismo , Interações Medicamentosas , Feminino , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/metabolismo , Masculino , Camundongos , Midazolam/administração & dosagem , Midazolam/sangue , Midazolam/metabolismo , Teratogênicos/metabolismo , Talidomida/administração & dosagem , Talidomida/metabolismo
8.
Actual. anestesiol. reanim ; 21(2): 27-30, abr.-jun. 2011. tab
Artigo em Espanhol | IBECS | ID: ibc-97921

RESUMO

En su conjunto, la respuesta inmune pretende: destruir los elementos extraños o no reparables del organismo; delimitar y aislar el foco inflamatorio, reparar las lesiones promoviendo la cicatrización y la neovascularización, activar los mecanismos generales que aporten células y nutrientes (activación neuroendocrina y metabólica); y evitar la generalización del proceso, induciendo una cierta inmunodepresión sistémica contrarreguladora. En el presente trabajo se hace una revisión sobre los aspectos fisiopatológicos derivados de la respuesta metabólica, inmunológica e inflamatoria a la agresión quirúrgica(AU)


On the Whole, the immune response seeks: to destroy the foreing or not repairable elements of the organism; to limit and to isolate the focus of the inflamation; to repair the lesion by advancyng the cicatrization and the neovascularización; to activate the general mechanisms that bring cells and nutrients (neuroendocrina and metabolic activation); and to avoid the generalization of the process inducing a certain systemic contraregulatory inmunodepression In the present work a review is maid of the physiopatologyc aspects derived from the metabolic, immunological and inflammatory response to the surgical aggression(AU)


Assuntos
Humanos , Anestesia/métodos , Anestesia/tendências , Relação Dose-Resposta a Droga , Relação Dose-Resposta Imunológica , Neovascularização Fisiológica , Neovascularização Fisiológica/imunologia , Adjuvantes Anestésicos/imunologia , Adjuvantes Anestésicos/metabolismo , Anestésicos/imunologia , Anestésicos/metabolismo , Inibidores da Angiogênese/imunologia , Anestésicos Gerais/imunologia , Anestésicos Gerais/metabolismo , Anestésicos Combinados/imunologia , Anestésicos Combinados/metabolismo
9.
Drug Metab Dispos ; 39(4): 703-10, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21177984

RESUMO

Ticagrelor is an orally administered, antiplatelet agent that inhibits the prothrombotic effects of ADP on the platelet by antagonizing the P2Y(12) receptor. Ticagrelor is a reversibly binding direct-acting P2Y(12) antagonist and does not require metabolic activation to achieve its antiplatelet effect. CYP3A4 and CYP3A5 appear to be the enzymes predominantly responsible for the formation of the ticagrelor active and inactive metabolites, AR-C124910XX and AR-C133913XX. The apparent K(m) values in human liver microsomes are 27.0 and 38.8 µM, with V(max) values of 730 and 417 pmol/min/mg for AR-C124910XX and AR-C133913XX, respectively. Ticagrelor moderately inhibited CYP2C9 activity in human liver microsomes with an IC(50) of 10.5 µM, while exhibiting little or no inhibition of CYP1A2, CYP2B6, CYP2C8, CYP2C19, CYP2D6, and CYP2E1. In human liver microsomes, ticagrelor inhibited midazolam 4-hydroxylation with an IC(50) of 8.2 µM, while activating 1'-hydroxylation of midazolam. Studies with recombinant enzymes suggested that cytochrome b(5) and CYP3A4 interactions play a significant role in this differential kinetic behavior. Evaluated in fresh human hepatocytes at concentration up to 20 µM, ticagrelor was not an inducer of CYP1A2 or CYP3A4. Although ticagrelor exhibited a tendency for CYP2B6 and CYP2C9 induction, its potential to cause drug interactions via the induction of these enzymes is low when its exposure at a therapeutic dose is considered.


Assuntos
Adenosina/análogos & derivados , Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/metabolismo , Microssomos Hepáticos/metabolismo , Inibidores da Agregação Plaquetária/metabolismo , Antagonistas do Receptor Purinérgico P2Y/metabolismo , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacocinética , Adjuvantes Anestésicos/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Hepatócitos/efeitos dos fármacos , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Midazolam/metabolismo , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacocinética , Antagonistas do Receptor Purinérgico P2Y/química , Antagonistas do Receptor Purinérgico P2Y/farmacocinética , Ticagrelor
10.
Planta ; 231(4): 977-90, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20108000

RESUMO

We previously showed that recombinant extra domain A from fibronectin (EDA) purified from Escherichia coli was able to bind to toll-like receptor 4 (TLR4) and stimulate production of proinflammatory cytokines by dendritic cells. Because EDA could be used as an adjuvant for vaccine development, we aimed to express it from the tobacco plastome, a promising strategy in molecular farming. To optimize the amount of recombinant EDA (rEDA) in tobacco leaves, different downstream sequences were evaluated as potential fusion tags. Plants generated by tobacco plastid transformation accumulated rEDA at levels up to 2% of the total cellular protein (equivalent to approximately 0.3 mg/g fresh weight) when translationally fused to the first 15 amino acids of green fluorescence protein (GFP). The recombinant adjuvant could be purified from tobacco leaves using a simple procedure, involving ammonium sulfate precipitation and anion exchange chromatography. Purified protein was able to induce production of tumour necrosis factor-alpha (TNF-alpha) either by bone marrow-derived dendritic cells or THP-1 monocytes. The rEDA produced in tobacco leaves was also able to induce upregulation of CD54 and CD86 maturation markers on dendritic cells, suggesting that the rEDA retains the proinflammatory properties of the EDA produced in E. coli and thus could be used as an adjuvant in vaccination against infectious agents and cancer. Taken together, these results demonstrate that chloroplasts are an attractive production vehicle for the expression of this protein vaccine adjuvant.


Assuntos
Adjuvantes Anestésicos/farmacologia , Cloroplastos/metabolismo , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Nicotiana/metabolismo , Plantas Geneticamente Modificadas/metabolismo , Adjuvantes Anestésicos/metabolismo , Animais , Northern Blotting , Southern Blotting , Células Cultivadas , Cloroplastos/genética , Cromatografia por Troca Iônica , Células Dendríticas/efeitos dos fármacos , Fibronectinas/genética , Camundongos , Plantas Geneticamente Modificadas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Nicotiana/genética
11.
Mol Diagn Ther ; 13(5): 331-7, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19791836

RESUMO

BACKGROUND: There is much variation in the response of both individuals and different ethnic populations to opioids, with genetic differences being responsible for interindividual variation. The micro-opioid receptor single nucleotide polymorphism (rs number 1799971) at nucleotide position 118 (OPRM1 118A>G) affects the analgesic response to opioids. OBJECTIVE: This study aimed to investigate the association between the OPRM1 118A>G polymorphism and the effects of fentanyl-induced analgesia, respiratory depression, and anesthetic recovery responses in a population of Han Chinese patients. STUDY DESIGN: The study was a case series in a hospital setting, with 1 year of study and 1 year of follow-up. A total of 189 patients (92 males and 97 females; American Society of Anesthesiologists Physical Status I or II, Glasgow Coma Scale = 15) who were scheduled for laparoscopic abdominal surgery received intravenous midazolam (Versed) 0.08-0.01 mg/kg and fentanyl (Duragesic) 5.0 microg/kg. The main outcome measure was the degree of postoperative pain, as assessed using the Visual Analog Scale (VAS). VAS scores were recorded 5, 15, 30, 45, and 60 minutes after a fentanyl bolus injection in the post-anesthesia care unit (PACU). The minute expiratory volume, end-tidal carbon dioxide concentration (EtCO(2)) and respiratory rate (RR) were measured continuously. The incidence of fentanyl-induced respiratory depression (RR <8/min and EtCO(2) >45 mmHg) was recorded at its appearance and treated with respiratory assistance. Blood gas analysis was done 15, 30, 45, and 60 minutes after extubation. These parameters were correlated with genotyping results of genomic DNA extracted from whole blood. RESULTS: Patients with the OPRM1 118 AG or GG genotypes had significantly higher VAS pain scores 15 and 30 minutes after a fentanyl bolus injection in the PACU than AA genotype patients (p < 0.05). A small but statistically significant difference was observed between the 118 AA and 118 AG/GG genotypes with regard to the carbon dioxide arterial pressure (PaCO(2)) at 15 and 30 minutes from the fentanyl bolus injection after extubation (p < 0.05); however, no clinically significant difference in the frequency of respiratory depression was seen. Homozygous 118 GG genotype patients had a significantly shorter time to awakening (p = 0.018) and extubation (p = 0.024) than patients with the 118 AA genotype. When the 118 GG and 118 AG genotypes were combined for analysis, a significantly shorter time to awakening (p = 0.011) and extubation (p = 0.010), compared with the 118 AA genotype, was also seen. CONCLUSION: The OPRM1 118A>G polymorphism lessens the analgesic response to fentanyl and the time to awakening and extubation but has no clinically significant effect on the incidence of respiratory depression.


Assuntos
Adjuvantes Anestésicos/metabolismo , Anestesia/métodos , Fentanila/metabolismo , Polimorfismo Genético , Receptores Opioides mu/genética , Adulto , Povo Asiático/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade
12.
Clin Cancer Res ; 12(24): 7492-7, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17189422

RESUMO

PURPOSE: Many chemotherapeutic drugs have an inherent lack of safety due to interindividual variability of hepatic cytochrome P450 (CYP) 3A4 drug metabolism. This reduction in CYP3A4 in cancer patients is possibly mediated by cytokines associated with tumor-derived inflammation. We sought to examine this link by using an explant sarcoma in a novel transgenic mouse model of human CYP3A4 regulation. EXPERIMENTAL DESIGN: Engelbreth-Holm-Swarm sarcoma cells were injected into the hindlimb of transgenic CYP3A4/lacZ mice. Hepatic expression of the human CYP3A4 transgene was analyzed by direct measurement of the reporter gene product, beta-galactosidase enzyme activity. Hepatic expression of murine Cyp3a was analyzed at the mRNA, protein, and function levels. The acute phase response was assessed by examining cytokines [interleukin-6 (IL-6) and tumor necrosis factor] in serum, liver, or tumor as well as hepatic expression of serum amyloid protein P. RESULTS: Engelbreth-Holm-Swarm sarcoma elicited an acute phase response that coincided with down-regulation of the human CYP3A4 transgene in the liver as well as the mouse orthologue Cyp3a11. The reduction of murine hepatic Cyp3a gene expression in tumor-bearing mice resulted in decreased Cyp3a protein expression and consequently a significant reduction in Cyp3a-mediated metabolism of midazolam. Circulating IL-6 was elevated and IL-6 protein was only detected in tumor tissue but not in hepatic tissue. CONCLUSIONS: The current study provides a mechanistic link between cancer-associated inflammation and impaired drug metabolism in vivo. Targeted therapy to reduce inflammation may provide improved clinical benefit for chemotherapy drugs metabolized by hepatic CYP3A4 by improving their pharmacokinetic profile.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/metabolismo , Sarcoma Experimental/metabolismo , Adjuvantes Anestésicos/metabolismo , Adjuvantes Anestésicos/farmacologia , Animais , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Midazolam/metabolismo , Midazolam/farmacologia , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Transgenes , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
13.
J Pharm Biomed Anal ; 41(4): 1332-41, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16621415

RESUMO

A sensitive enzyme-linked immunosorbent assay (ELISA) was developed for the detection of fentanyl in serum and urine. The ELISA used an indirect competitive method produced by coating the plate with thyroglobulin conjugated with fentanyl hapten. Antibodies against fentanyl-hemocyanin were detected by a goat-anti-rabbit antibody conjugated with alkaline phosphatase. Calibration standard curves ranged from 0.5ng/ml to 50mug/ml (IC(50)=10ng/ml), and the limits of detection were 0.5 and 1.0ng/ml for serum and urine, respectively. The intra- and inter-assay variations were less than 8% and 10%, respectively. The antibody produced against fentanyl completely cross-reacted with p-fluorofentanyl, thienylfentanyl and 3-methylthienylfentanyl, cross-reacted highly with carfentanil (85%), but was considered non-cross-reactive with alpha-methylfentanyl (5%), sufentanil (<1%), alfentanil (<1%) and lofentanil (<1%). Nano-sized iron oxide magnetic particles coated with the developed fentanyl antibody were capable of specific binding and releasing of fentanyl from urine samples. This enabled the drug to be effectively pre-concentrated and decreased the limit of detection by approximately one order of magnitude. The analytical background noise was significantly reduced to enable fentanyl detection at concentrations originally below chromatographic limit of detection. The change of platform for antibody binding with nanoparticles demonstrated a novel use of antibodies for sample preparation and should facilitate drug screening by traditional ELISA.


Assuntos
Adjuvantes Anestésicos/urina , Anticorpos/metabolismo , Fentanila/urina , Adjuvantes Anestésicos/sangue , Adjuvantes Anestésicos/metabolismo , Animais , Antibacterianos/urina , Reações Cruzadas , Eletroforese Capilar/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Fentanila/análogos & derivados , Fentanila/metabolismo , Cavalos , Nanoestruturas , Coelhos
14.
Biochem Pharmacol ; 66(12): 2333-40, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14637191

RESUMO

Effects of cytochrome b(5) (b(5)) on catalytic activities of human cytochrome P450 (CYP) 3A5, CYP3A4, and CYP3A7 coexpressed with human NADPH-cytochrome P450 reductase in Escherichia coli membranes were investigated using 14 substrates. The activities of CYP3A5 were enhanced by addition of b(5) in approximately one third of the substrates employed in this study. Such enhancement by b(5) was roughly similar to that of CYP3A4, while the activities of CYP3A7 were not enhanced by b(5) with any substrates employed. V(max) values for midazolam 1'-hydroxylation and amitriptyline N-demethylation by CYP3A5 were increased about twice by addition of b(5), which was also seen with CYP3A4, although the extent of the effects of b(5) on S(50) (K(m)) and Hill coefficient differed dependent on substrates used. In contrast, b(5) did not alter any of these kinetic parameters of CYP3A7. The effects of b(5) on kinetic parameters of CYP3A5 were similar to those of CYP3A4 but not CYP3A7. These results suggest that roles of b(5) in drug oxidation activities of CYP3A5 and CYP3A4 are different from those of CYP3A7.


Assuntos
Amitriptilina/metabolismo , Hidrocarboneto de Aril Hidroxilases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Citocromos b5/farmacologia , Midazolam/metabolismo , Adjuvantes Anestésicos/metabolismo , Analgésicos não Narcóticos/metabolismo , Hidrocarboneto de Aril Hidroxilases/genética , Catálise , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Humanos , Cinética , Oxirredução , Homologia de Sequência de Aminoácidos
15.
Eur J Clin Pharmacol ; 58(10): 683-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12610745

RESUMO

OBJECTIVE: NN703 (tabimorelin) is an orally active growth hormone (GH) secretagogue intended for use as an alternative to daily injections of GH. In vitro studies in human liver microsomes have indicated that NN703 is a mechanism-based inhibitor of CYP3A4. The aim of the present study was to investigate in man the effects of NN703 on the pharmacokinetics of midazolam, a substrate of CYP3A4. METHODS: Seventeen adult male subjects were enrolled in the study, and each received an oral dose of midazolam (7.5 mg) on four occasions: at baseline (day 1), after one dose of NN703 (day 3), after 7 days once daily NN703 treatment (day 9) and after a 7-day washout period (day 16). The pharmacokinetics of midazolam and its main metabolite, alpha-hydroxymidazolam, were investigated. RESULTS: Following a single dose of NN703 (day 3), the AUC of both midazolam and alpha-hydroxymidazolam increased by 64% and 34%, respectively (P=0.0001 for both). After repeated NN703 dosing (day 9), NN703 levels reached steady state, and midazolam AUC further increased to 93% relative to baseline (P=0.0001), whereas alpha-hydroxymidazolam AUC decreased slightly and was 11% higher than baseline (n.s.). Following the washout period (day 16), midazolam AUC decreased to values lower than those on day 3 and day 9, but still significantly (45%) higher than baseline levels (P=0.0001). The C(max) values of midazolam and alpha-hydroxymidazolam demonstrated a pattern similar to the AUC, but the effect following repeated NN703 dosing was more pronounced. The t(1/2) of midazolam increased from day 1 to day 3 (P=0.0483) but was essentially unchanged at steady state on day 9. CONCLUSION: This study shows that administration of NN703 and midazolam, a CYP3A4 substrate, leads to a significant increase in exposure of midazolam. This is consistent with NN703 inhibition of CYP3A4 activity.


Assuntos
Adjuvantes Anestésicos/farmacocinética , Inibidores das Enzimas do Citocromo P-450 , Dipeptídeos/farmacologia , Midazolam/análogos & derivados , Midazolam/farmacocinética , Adjuvantes Anestésicos/metabolismo , Adulto , Área Sob a Curva , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Dipeptídeos/efeitos adversos , Interações Medicamentosas , Meia-Vida , Humanos , Masculino , Midazolam/metabolismo , Pessoa de Meia-Idade , Fatores de Tempo
16.
Biochem Pharmacol ; 64(4): 633-43, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12167482

RESUMO

The cytochrome P450 3A (CYP3A)-mediated midazolam oxidation was studied in rat precision-cut liver slices (PCLS) maintained for 20hr at 4, 20 and 37 degrees, and further incubated for 8hr at 37 degrees. Either at 4 or 20 degrees, midazolam was oxidised by PCLS at similar rates to that observed in freshly cut slices. Moreover, PCLS kept a regioselectivity since 4-hydroxylation was more important than 1'-hydroxylation. Conversely, PCLS totally lost their capacity to oxidise midazolam after 20hr at 37 degrees, and both CYP3A2 protein and mRNA were not detected. CYP3A1 protein was unaffected by a temperature of 37 degrees but its mRNA was totally lost. By blocking transcription with actinomycin D, the decay of both CYP3A mRNAs followed the same profile at either 20 or 37 degrees, indicating that temperature affected the CYP3A2 protein stability. Cell functionality was not involved in such an impairment since the low values of ATP, GSH and protein synthesis rates observed at 4 and 20 degrees were rapidly restored, when PCLS were further incubated at 37 degrees. The use of rat supersomes expressing either CYP3A1 or CYP3A2, strongly supported the hypothesis that 4-hydroxymidazolam was mainly formed by CYP3A2. These results suggest that: (1) CYP3A1 protein is constitutive and largely expressed in rat liver slices; (2) regioselective midazolam oxidation appears to be mainly CYP3A2 dependent; and (3) since CYP3A isoforms have similar half-lives (about 10-14hr), the loss of CYP3A2 protein at 37 degrees might be due to a selective targeting (phosphorylation ?) leading to proteolytic disposal by the proteasome.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/biossíntese , Isoenzimas/biossíntese , Fígado/enzimologia , Midazolam/metabolismo , Oxirredutases N-Desmetilantes/biossíntese , Trifosfato de Adenosina/metabolismo , Adjuvantes Anestésicos/metabolismo , Animais , Western Blotting , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Glutationa/metabolismo , Técnicas In Vitro , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/metabolismo , Masculino , Oxirredução , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Temperatura
17.
Ann Emerg Med ; 31(6): 729-36, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9624313

RESUMO

gamma-Hydroxybutyric acid (GHB) is unfamiliar to many physicians in the United States but enjoys clinical use elsewhere for applications in resuscitation, anesthesia, and addiction therapy. Use within the United States is restricted to Food and Drug Administration-approved clinical trials for treatment of narcolepsy. Recently illicit use of GHB has emerged within the United States where it is distributed for purported euphoric and "fat-burning" metabolic effects. Clinical effects can be severe, progressing rapidly to respiratory arrest and death. We provide an updated comprehensive review of the literature with particular emphasis on toxicology, including GHB pharmacodynamics, clinical effects, and suggestions for overdose management. Recommended management of acute GHB intoxication includes prevention of aspiration, use of atropine for persistent symptomatic bradycardia, consideration of neostigmine as a reversal agent, and treatment for coingested substances. Emergency physicians are urged to become familiar with GHB because of its potential for severe morbidity as well as its potential use as a future resuscitative agent.


Assuntos
Adjuvantes Anestésicos/intoxicação , Oxibato de Sódio/intoxicação , Adjuvantes Anestésicos/metabolismo , Adjuvantes Anestésicos/farmacologia , Animais , Overdose de Drogas/terapia , Humanos , Oxibato de Sódio/metabolismo , Oxibato de Sódio/farmacologia , Transtornos Relacionados ao Uso de Substâncias
19.
Acta Anaesthesiol Belg ; 35 Suppl: 399-411, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-6516748

RESUMO

When suitable, local anesthetic techniques are preferable for ambulatory short diagnostic procedures. If general anesthesia is used no or only very light premedication, induction with a short acting i.v. agent and the maintenance of anesthesia with nitrous oxide plus oxygen plus an inhalational agent or alternatively with short acting i.v. anesthetic and analgesic seems today to provide fastest recovery. Patients should always arrange for a responsible adult to accompany them home. Minimal requirements for safe discharge should in addition to stable vital signs and the ability to maintain oral fluids include that patients are able to dress themselves and able to walk out. These criteria, however, do not indicate that patients are fully recovered after anesthesia. In most cases patients are not fully recovered when discharged from hospital after even brief anesthesia and they should refrain from driving, operating machinery, and delay making important decisions for at least 24 hours after anesthesia.


Assuntos
Assistência Ambulatorial , Anestesia/métodos , Alta do Paciente , Adjuvantes Anestésicos/metabolismo , Anestésicos/metabolismo , Humanos , Desempenho Psicomotor
20.
Acta Anaesthesiol Scand ; 23(3): 285-90, 1979 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-484185

RESUMO

To investigate the effect of different anesthetics on the pharmacokinetics and pharmacodynamics of pancuronium, 120 microgram/kg i.v., cats were anesthetized with either pentobarbital (N = 4), ketamine (N = 4), enflurane (N = 5), or halothane (N = 5). A longer onset time and duration of neuromuscular blockade occurred during enflurane and halothane anesthesia. The apparent elimination half-life was longer and the total voluem of distribution at steady state larger during halothane anesthesia. The plasma concentration of pancuronium required for neuromuscular blockades was less during enflurane than during the other three anesthetics. We conclude that inhalation anesthetics may prolong a neuromuscular blockade by altering both the pharmacokinetics and pharmacodynamics of pancuronium.


Assuntos
Adjuvantes Anestésicos/metabolismo , Anestésicos/farmacologia , Junção Neuromuscular/efeitos dos fármacos , Pancurônio/metabolismo , Adjuvantes Anestésicos/administração & dosagem , Adjuvantes Anestésicos/farmacologia , Animais , Gatos , Enflurano/farmacologia , Halotano/farmacologia , Ketamina/farmacologia , Pancurônio/administração & dosagem , Pancurônio/farmacologia , Pentobarbital/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...