Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Pharmacol Ther ; 102(3): 436-449, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28599065

RESUMO

Recent advances in the identification and characterization of renal drug transporters and drug-metabolizing enzymes has led to greater understanding of their roles in drug and chemical elimination and in modulation of the intrarenal exposure and response to drugs, nephrotoxic compounds, and physiological mediators. Furthermore, there is increasing awareness of the potential importance of drug-drug interactions (DDIs) arising from inhibition of renal transporters, and regulatory agencies now provide recommendations for the evaluation of transporter-mediated DDIs. Apart from the well-recognized effects of kidney disease on renal drug clearance, there is a growing body of evidence demonstrating that the nonrenal clearances of drugs eliminated by certain transporters and drug-metabolizing enzymes are decreased in patients with chronic kidney disease (CKD). Based on these observations, renal impairment guidance documents of regulatory agencies recommend pharmacokinetic characterization of both renally cleared and nonrenally cleared drugs in CKD patients to inform possible dosage adjustment.


Assuntos
Nefropatias/fisiopatologia , Rim/metabolismo , Preparações Farmacêuticas/metabolismo , Animais , Transporte Biológico , Interações Medicamentosas , Humanos , Rim/fisiopatologia , Nefropatias/metabolismo , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/fisiopatologia
2.
Drug Metab Dispos ; 35(3): 363-70, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17151189

RESUMO

The stereo- and regioselective glucuronidation of 10 Delta(4)-3-keto monohydroxylated androgens and pregnanes was investigated to identify UDP-glucuronosyltransferase (UGT) enzyme-selective substrates. Kinetic studies were performed using human liver microsomes (HLMs) and a panel of 12 recombinant human UGTs as the enzyme sources. Five of the steroids, which were hydroxylated in the 6beta-, 7alpha-, 11beta- or 17alpha-positions, were not glucuronidated by HLMs. Of the remaining compounds, comparative kinetic and inhibition studies indicated that 6alpha- and 21-hydroxyprogesterone (OHP) were glucuronidated selectively by human liver microsomal UGT2B7. 6alpha-OHP glucuronidation by HLMs and UGT2B7 followed Michaelis-Menten kinetics, whereas 21-OHP glucuronidation by these enzyme sources exhibited positive cooperativity. UGT2B7 was also identified as the enzyme responsible for the high-affinity component of human liver microsomal 11alpha-OHP glucuronidation. In contrast, UGT2B15 and UGT2B17 were the major forms involved in human liver microsomal testosterone 17beta-glucuronidation and the high-affinity component of 16alpha-OHP glucuronidation. Activity of UGT1A subfamily enzymes toward the hepatically glucuronidated substrates was generally low, although UGT1A4 and UGT1A9 contribute to the low-affinity components of microsomal 16alpha- and 11alpha-OHP glucuronidation, respectively. Interestingly, UGT1A10, which is expressed only in the gastrointestinal tract, exhibited activity toward most of the glucuronidated substrates. The results indicate that 6alpha- and 21-OHP may be used as selective "probes" for human liver microsomal UGT2B7 activity and, taken together, provide insights into the regio- and stereoselectivity of hydroxysteroid glucuronidation by human UGTs.


Assuntos
Glucuronídeos/biossíntese , Glucuronosiltransferase/metabolismo , Hidroxiprogesteronas/metabolismo , Linhagem Celular , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/genética , Humanos , Microssomos Hepáticos/metabolismo , Proteínas Recombinantes/metabolismo , Testosterona/metabolismo , Zidovudina/farmacologia
3.
Arch Biochem Biophys ; 396(1): 56-64, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11716462

RESUMO

Acyl-CoA conjugation of xenobiotic carboxylic acids is catalyzed by hepatic microsomal long-chain fatty acid CoA ligases (LCL, EC 6.2.1.3). Marmosets (Callithrix jacchus) are considered genetically closer to humans than rodents and are used in pharmacological and toxicological studies. We have demonstrated that marmoset liver microsomes catalyze nafenopin-, ciprofibroyl-, and palmitoyl-CoA conjugation and that only palmitoyl-CoA conjugation is significantly upregulated (1.7-fold, P < 0.02) by a high fat diet. Additionally, the apparent C(50) values for nafenopin-, ciprofibroyl-, and palmitoyl-CoA conjugation of 149.7, 413.4, and 3.4 microM were comparable to those reported for human liver microsomes viz, 213.7, 379.8, and 3.4 microM, respectively. Comparison with human data was enabled by the cloning of a full-length marmoset cDNA (MLCL1) that encoded a 698-amino-acid protein sharing 83% similarity with rat liver acyl-CoA synthetase (ACS1) and 93 and 90% similarity with human liver LCL1 and LCL2, respectively. MLCL1 transiently expressed in COS-7 cells activated nafenopin (C(50) 192.9 microM), ciprofibrate (C(50) 168.7 microM), and palmitic acid (C(50) 4.5 microM) to their respective CoA conjugates. This study also demonstrated that the sigmoidal kinetics observed for nafenopin- and ciprofibroyl-CoA conjugation were not unique to human liver microsomes but were also characteristic of marmoset liver microsomes and recombinant MLCL1. More extensive characterization of the substrate specificity of marmoset LCL isoforms will aid in determining further the suitability of marmosets as a model for human xenobiotic metabolism via acyl-CoA conjugation.


Assuntos
Callithrix/genética , Ácido Clofíbrico/análogos & derivados , Ácido Clofíbrico/metabolismo , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Microssomos Hepáticos/metabolismo , Nafenopina/metabolismo , Ácido Palmítico/metabolismo , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Sequência de Aminoácidos , Animais , Ácidos Fíbricos , Humanos , Masculino , Dados de Sequência Molecular , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Xenobióticos/metabolismo
4.
Biochem Pharmacol ; 60(4): 507-16, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10874125

RESUMO

The aromatic retinoid acitretin is the primary active metabolite of etretinate, and in this study we investigated the ethyl esterification of acitretin to etretinate using [(14)C]acitretin and human liver microsomes. Samples were analysed by TLC, HPLC, and LC-MS. Essential requirements for the transesterification reaction were identified and included viable microsomal protein, ATP, CoASH, and ethanol. Human liver microsomes catalysed formation of acitretinoyl-CoA at the rate of 0.08 +/- 0.02 nmol/min/mg (mean +/- SD, N = 10). Acitretinoyl-CoA was pivotal for the transesterification to etretinate and in the presence of methanol, ethanol, n-propanol, n-butanol, and hexanol, the corresponding esters, namely methyl-, ethyl (etretinate)-, propyl-, butyl-, and hexyl-acitretinate, were formed. On average, 1.7% of the acitretin present in the incubation was converted to etretinate in the presence of ethanol. In the absence of ethanol, transesterification did not proceed. Inhibition of the ester hydrolysis of etretinate by bis-p-nitrophenylphosphate (BNPP, 1 mM) prevented futile cycling of etretinate via acitretinoyl-CoA. An additional finding was that acitretin (15-30 microM) activated significantly human liver microsomal long-chain fatty acid-CoA ligase (E.C.6.2.1.3, LCL), resulting in enhanced formation of palmitoyl-CoA. This study demonstrated that in the presence of ethanol the ethyl esterification of acitretin to etretinate proceeds via formation of acitretinoyl-CoA. Predicting clearance of acitretin in vivo via this unique metabolic pathway will be a challenge, as the intracellular concentration of ethanol could never be predicted with any degree of accuracy in humans.


Assuntos
Acitretina/metabolismo , Etretinato/metabolismo , Microssomos Hepáticos/metabolismo , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Acetaldeído/metabolismo , Acetatos/metabolismo , Acil Coenzima A/análise , Coenzima A Ligases/metabolismo , Esterificação , Etanol/metabolismo , Humanos , Técnicas In Vitro
5.
Toxicol Appl Pharmacol ; 163(2): 176-82, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10698675

RESUMO

Endogenous fatty acyl-CoAs play an important role in the acylation of proteins. A number of xenobiotic carboxylic acids are able to mimic fatty acids, forming CoA conjugates and acting as substrates in pathways of lipid metabolism. In this study nafenopin, a substrate for human hepatic fatty acid-CoA ligases, was chosen as a model compound to study xenobiotic acylation of human liver proteins. (3)H-nafenopin (+/- unlabeled palmitate) or (14)C-palmitate (+/- unlabeled nafenopin) were incubated for up to 120 min at 37 degrees C with ATP, CoA, and homogenate protein (1 mg/ml) from four individual human livers. Nafenopin covalently bound to proteins was detectable in all human livers and increased with time. Nafenopin adduct formation was directly proportional to nafenopin-CoA formation (r = 0.985, p < 0.05). Attachment of nafenopin to proteins involved both thioester and amide linkages with 76 and 24% of adducts formed with proteins > 100 and 50-100 kDa, respectively. Protein acylation by palmitate was also demonstrated. Palmitate significantly inhibited nafenopin-CoA formation by 29% but had no effect on nafenopin-CoA-mediated protein acylation. In contrast, nafenopin significantly inhibited protein palmitoylation by palmitoyl-CoA. This is the first study to demonstrate a direct relationship between xenobiotic-CoA formation, acylation of human liver proteins, and inhibition of endogenous palmitoylation. The ability of xenobiotics to acylate tissue proteins may have important biological consequences including perturbation of endogenous regulation of protein localization and function.


Assuntos
Acil Coenzima A/metabolismo , Fígado/metabolismo , Nafenopina/análogos & derivados , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Acil Coenzima A/química , Acilação , Adolescente , Adulto , Amidas/química , Coenzima A Ligases/metabolismo , Feminino , Humanos , Técnicas In Vitro , Fígado/química , Masculino , Pessoa de Meia-Idade , Nafenopina/química , Nafenopina/metabolismo , Palmitatos/metabolismo , Proteínas/química , Compostos de Sulfidrila/química , Xenobióticos/química
6.
Curr Drug Metab ; 1(1): 49-66, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11467080

RESUMO

This review focuses primarily on the mammalian medium and long-chain fatty acid coenzyme A ligases that have been implicated in the metabolism of xenobiotic carboxylic acids such as pesticides, arylpropionate non steroidal anti-inflammatory drugs and the hypolipidaemic clofibrate and its congeners. Evidence of multiplicity of mitochondrial and microsomal enzymes and their respective substrate/inhibitor profiles are discussed. For completeness, where appropriate, details of non-substrate inhibitors have also been included. Although knowledge is limited at present with respect to the medium-chain enzymes, aspects of regulation particularly the in vivo, in vitro role of peroxisome proliferators and current knowledge of the molecular biology of the long-chain fatty acid CoA ligase superfamily are documented. Additionally, alignment of thirteen cloned mammalian fatty acid CoA ligases using criteria established for the CYP and UGT superfamilies has enabled construction of a phylogenetic tree that clearly defines three families. Catalytic data are still limited and the xenobiotic substrate/inhibitor profiles of the recombinant proteins are incomplete. Finally, with increasing recognition of the importance of fatty acyl-CoA esters as physiological regulators of cell function including gene expression, the review concludes with a discussion of the metabolic fate and toxicity of xenobiotic acyl-CoA esters.


Assuntos
Coenzima A/metabolismo , Ácidos Graxos/metabolismo , Ligases/metabolismo , Xenobióticos/metabolismo , Animais , Coenzima A/química , Humanos , Cinética , Ligases/química , Preparações Farmacêuticas/metabolismo
7.
Clin Exp Pharmacol Physiol ; 25(10): 776-82, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9784915

RESUMO

1. Formation of acyl-coenzymes (Co)A occurs as an obligatory step in the metabolism of a variety of endogenous substrates, including fatty acids. The reaction is catalysed by ATP-dependent acid:CoA ligases (EC 6.2.1.1-2.1.3; AMP forming), classified on the basis of their ability to conjugate saturated fatty acids of differing chain lengths, short (C2-C4), medium (C4-C12) and long (C10-C22). The enzymes are located in various cell compartments (cytosol, smooth endoplasmic reticulum, mitochondria and peroxisomes) and exhibit wide tissue distribution, with highest activity associated with liver and adipose tissue. 2. Formation of acyl-CoA is not unique to endogenous substrates, but also occurs as an obligatory step in the metabolism of some xenobiotic carboxylic acids. The mitochondrial medium-chain CoA ligase is principally associated with metabolism via amino acid conjugation and activates substrates such as benzoic and salicylic acids. Although amino acid conjugation was previously considered an a priori route of metabolism for xenobiotic-CoA, it is now recognized that these highly reactive and potentially toxic intermediates function as alternative substrates in pathways of intermediary metabolism, particularly those associated with lipid biosyntheses. 3. In addition to a role in fatty acid metabolism, the hepatic microsomal and peroxisomal long-chain-CoA-ligases have been implicated in the formation of the acyl-CoA thioesters of a variety of hypolipidaemic and peroxisome proliferating agents (e.g. clofibric acid) and of the R(-)-enantiomers of the commonly used 2-arylpropionic acid non-steroidal anti-inflammatory drugs (e.g. ibuprofen). In vitro kinetic studies using rat hepatic microsomes and peroxisomes have alluded to the possibility of xenobiotic-CoA ligase multiplicity. Although cDNA encoding a long-chain ligase have been isolated from rat and human liver, there is currently no molecular evidence of multiple isoforms. The gene has been localized to chromosome 4 and homology searches have revealed a significant similarity with enzymes of the luciferase family. 4. Increasing recognition that formation of a CoA conjugate increases chemical reactivity of xenobiotic carboxylic acids has led to an awareness that the relative activity, substrate specificity and intracellular location of the xenobiotic-CoA ligases may explain differences in toxicity. 5. Continued characterization of the human xenobiotic-CoA ligases in terms of substrate/inhibitor profiles and regulation, will allow a greater understanding of the role of these enzymes in the metabolism of carboxylic acids.


Assuntos
Ácidos Carboxílicos/metabolismo , Coenzima A Ligases/metabolismo , Fígado/enzimologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Xenobióticos/metabolismo , Xenobióticos/toxicidade , Acetato-CoA Ligase/genética , Acetato-CoA Ligase/metabolismo , Animais , Coenzima A Ligases/genética , Humanos , Especificidade por Substrato
8.
Br J Clin Pharmacol ; 40(2): 153-6, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8562298

RESUMO

The effects of gender and oral contraceptive steroids on the pharmacokinetics of (R)-ibuprofen were studied in groups of healthy adult males, females and oral contraceptive steroid (OCS) using females. The values of AUC, CLpo, t1/2 and Vss, app did not differ significantly between the groups. Similarly, the percentage unbound of (R)-ibuprofen in pooled plasma from the three groups was not statistically different. Since chiral inversion is the major determinant of (R)-ibuprofen clearance in humans, it may be inferred from these data that gender and OCS have little or no effect on conversion of (R)-ibuprofen to the pharmacologically active S-enantiomer. Moreover, it is unlikely that hormonal factors influence the activity of the human hepatic long-chain fatty-acid:CoA ligase, the enzyme mediating the rate limiting step of (R)-ibuprofen inversion.


PIP: In Australia, clinical researchers studied the effects of gender and oral contraceptive (OC) steroids on the pharmacokinetics of (R)-ibuprofen in 8 healthy adult males (mean age = 21 years), adult females (24 years), and OC users (22 years). There were no statistically significant differences between males, females, and OC users for any pharmacokinetic parameter for (R)-ibuprofen. These parameters included areas under the plasma total concentration-time curve (AUC), maximal plasma concentration, time to maximal plasma concentration, half-life, CLpo, and apparent steady-state volumes of distribution. The AUC to the last data point observed for (S)-ibuprofen (derived from (R)-ibuprofen) was similar, suggesting that hormonal factors do not affect plasma clearance of (S)-ibuprofen. The average percentages unbound of (R)- and (S)-ibuprofen across the concentration was not statistically different between the groups: 1.82% and 2.84% for males, 1.83% and 3.01% for females, and 2.1% and 2.97% for OC users, respectively. The mean fraction unbound of (S)-ibuprofen was 53.6% greater than that of (R)-ibuprofen. Since chiral inversion may explain 62-92% of (R)-ibuprofen clearance in humans, these data may suggest that gender and OCs do not effect or have only a limited effect on the conversion of (R)-ibuprofen to the pharmacologically active S-enantiomer. These findings indicate that hormonal factors probably do not affect the activity of the human hepatic long-chain fatty-acid:CoA ligase, the enzyme mediating the rate limiting step of (R)-ibuprofen inversion.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Anticoncepcionais Orais Hormonais/farmacologia , Ibuprofeno/farmacocinética , Administração Oral , Adolescente , Adulto , Análise de Variância , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/sangue , Feminino , Humanos , Ibuprofeno/administração & dosagem , Ibuprofeno/sangue , Masculino , Fatores Sexuais , Estereoisomerismo
9.
Xenobiotica ; 25(5): 469-76, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7571720

RESUMO

1. Activity of rat hepatic microsomal and peroxisomal long-chain (palmitoyl) and nafenopin-CoA ligases were studied following administration of either clofibric acid, di-(2-ethylhexyl)phthalate (DEHP) or phenobarbitone. 2. Clofibric acid significantly induced the peroxisomal palmitoyl and nafenopin-CoA ligases, whilst no induction of the equivalent enzymes was observed in the microsomal fraction. 3. DEHP induced only palmitoyl-CoA formation in peroxisomes, whilst all enzymes were refractory to phenobarbitone treatment. 4. The enzyme-specific patterns of inductions both intra- and inter-organelle suggest that the palmitoyl and nafenopin-CoA ligases are under different regulatory control. 5. Modulation of both the rate and extent of nafenopin- and palmitoyl-CoA formation was both agent and organelle specific. 6. This study highlights the difficulty in delineating the individual roles of both fatty acyl-CoAs and xenobiotic-CoAs in peroxisome proliferation.


Assuntos
Ácido Clofíbrico/farmacologia , Coenzima A Ligases/biossíntese , Dietilexilftalato/farmacologia , Microcorpos/enzimologia , Microssomos Hepáticos/enzimologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Acil Coenzima A/metabolismo , Animais , Técnicas In Vitro , Masculino , Microcorpos/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Fenobarbital/farmacologia , Ratos , Ratos Sprague-Dawley
10.
Biochem Pharmacol ; 49(9): 1335-9, 1995 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-7763316

RESUMO

In this study we have demonstrated that rat hepatic peroxisomes catalyse the formation of nafenopin-CoA. The process is mediated by apparent high affinity (Km 6.7 microM), low capacity (Vmax 0.31 nmol/mg/min) and low affinity, high capacity isoforms. Palmitic acid (Ki 1.1 microM), R(-) ibuprofen (Ki 7.9 microM), ciprofibrate (Ki 60.2 microM) and clofibric acid (Ki 86.8 microM) competitively inhibited nafenopin-CoA formation catalysed by the apparent high affinity isoform. An antibody raised against the microsomal palmitoyl-CoA ligase inhibited the equivalent peroxisomal enzyme significantly (P < 0.001) but did not inhibit peroxisomal nafenopin-CoA ligase activity. These data suggest that nafenopin-CoA formation is catalysed by a peroxisomal CoA ligase which differs from the peroxisomal long chain fatty acid-CoA ligase in relation to its xenobiotic/antibody inhibitor profile and kinetic characteristics.


Assuntos
Coenzima A Ligases/metabolismo , Fígado/enzimologia , Microcorpos/enzimologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Animais , Ácido Clofíbrico/análogos & derivados , Ácido Clofíbrico/farmacologia , Coenzima A Ligases/antagonistas & inibidores , Ácidos Fíbricos , Ibuprofeno/farmacologia , Cinética , Masculino , Ácido Palmítico , Ácidos Palmíticos/farmacologia , Ratos , Ratos Sprague-Dawley
11.
Chem Biol Interact ; 90(3): 215-23, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8168170

RESUMO

Multiplicity of hepatic microsomal coenzyme A ligases catalyzing acyl-CoA thioester formation is an important factor for consideration in relation to the metabolism of xenobiotic carboxylic acids. In this study the kinetic characteristics of rat hepatic microsomal nafenopin-CoA ligase were studied and compared with those of long-chain fatty acid (palmitoyl) CoA ligase. The high affinity component of palmitoyl-CoA formation was inhibited by nafenopin (Ki 53 microM) and ciprofibrate (Ki 1000 microM). Analagous to palmitoyl-CoA, nafenopin-CoA formation was catalyzed by an apparent high affinity low capacity isoform (Km 6 +/- 2.5 microM, Vmax 0.33 +/- 0.12 nmol/mg per min) which was inhibited competitively by palmitic acid (mean Ki 1.7 microM, n = 5) and R-ibuprofen (mean Ki 10.8 microM, n = 5) whilst ciprofibrate and clofibric acid were ineffective as inhibitors. The intrinsic metabolic clearance of nafenopin to nafenopin-CoA (Vmax/Km 0.057 +/- 0.011 nmol/mg/min/ +/- M) was similar to that reported recently for the formation of ibuprofenyl-CoA by rat liver microsomes. Evidence of both a substantial difference between the Km and Ki for nafenopin and lack of commonality with regard to xenobiotic inhibitors suggests that the high affinity microsomal nafenopin-CoA and long-chain fatty acid-CoA ligases are kinetically distinct. Thus until the current 'long-chain like' xenobiotic-CoA ligases are fully characterised in terms of substrate specificity, inhibitor profile, etc, it will be impossible to rationalize (and possibly predict) the metabolism and hence toxicity of xenobiotic carboxylic acids forming acyl-CoA thioester intermediates.


Assuntos
Coenzima A Ligases/metabolismo , Microssomos Hepáticos/enzimologia , Nafenopina/metabolismo , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Acil Coenzima A/metabolismo , Animais , Ligação Competitiva , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Nafenopina/análogos & derivados , Nafenopina/farmacologia , Ácido Palmítico , Ácidos Palmíticos/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato , Xenobióticos/metabolismo
12.
Biochem Pharmacol ; 44(12): 2415-7, 1992 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-1472108

RESUMO

This study has demonstrated the involvement of multiple forms of rat hepatic microsomal CoA ligases in the formation of 2-arylpropionyl-CoA thioesters. In the presence of (-)R-ibuprofen (0.1 microM-1 mM) two enzymic processes were observed, one of which exhibited enantiospecificity and apparent high affinity for the R enantiomer (Km 0.06 microM) whilst the second, a low-affinity component was non-enantiospecific. An equivalent high-affinity isoform catalysing R-flurbiprofen-CoA formation at concentrations less than 100 microM was not demonstrated. However, at higher substrate concentrations formation of both R- and S-flurbiprofenyl-CoA thioesters occurred. Marked inter-individual variation was observed in the formation of S-ibuprofen-CoA and S-flurbiprofen-CoA in the rats studied.


Assuntos
Coenzima A Ligases/metabolismo , Isoenzimas/metabolismo , Microssomos Hepáticos/enzimologia , Tioléster Hidrolases/metabolismo , Animais , Ésteres/metabolismo , Flurbiprofeno/metabolismo , Ibuprofeno/metabolismo , Ratos , Estereoisomerismo , Compostos de Sulfidrila/metabolismo
13.
Biochem Pharmacol ; 44(7): 1291-6, 1992 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-1417953

RESUMO

In vivo and in vitro (-)R-ibuprofen is inverted to the (+)S antipode via stereoselective formation of an R-ibuprofenyl-CoA intermediate. In this study the effects of (-)R- and (+)S-ibuprofen on metabolism and respiration were studied using isolated rat hepatocytes and mitochondria. R-Ibuprofen significantly increased the lactate to pyruvate ratio, perturbed mitochondrial ketogenesis as evidenced by alterations in the beta-hydroxybutyrate to acetoacetate ratio and uncoupled mitochondrial oxidative phosphorylation. In addition, substantial dose- and time-dependent sequestration of reduced CoA (CoASH) occurred in the presence of the R enantiomer. Similarly, S-ibuprofen altered both the cytosolic and mitochondrial redox states although the magnitude of the effect was substantially less than that observed with the R enantiomer. In contrast to R-ibuprofen, S-ibuprofen did not uncouple oxidative phosphorylation or sequester hepatocyte CoASH. It is proposed that the perturbations observed in hepatocyte intermediary metabolism and mitochondrial function are attributable to a combination of the direct effects of R-ibuprofen per se and the sequestration of CoASH as R-ibuprofenyl-CoA during the process of chiral inversion. On the basis of these results, R-ibuprofen should be considered more in terms of metabolism to a reactive acyl-CoA intermediate rather than as a pro-drug for the pharmacologically active S-enantiomer.


Assuntos
Ibuprofeno/farmacologia , Fígado/efeitos dos fármacos , Ácido 3-Hidroxibutírico , Acetoacetatos/metabolismo , Animais , Hidroxibutiratos/metabolismo , Corpos Cetônicos/metabolismo , Lactatos/metabolismo , Ácido Láctico , Fígado/metabolismo , Masculino , Mitocôndrias Hepáticas/efeitos dos fármacos , Fosforilação Oxidativa , Consumo de Oxigênio , Piruvatos/metabolismo , Ácido Pirúvico , Ratos , Ratos Wistar , Estereoisomerismo
14.
Biochem Pharmacol ; 44(2): 261-7, 1992 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-1386510

RESUMO

ATP-dependent coenzyme A (CoA) ligases catalyse the formation of the acyl-CoA thioesters of xenobiotic carboxylic acids and the formation of xenobiotic-CoAs has been implicated as being a causative factor in peroxisomal proliferation. In this study we have demonstrated using rat liver peroxisomes that the formation of palmitoyl-CoA is inhibited by a variety of xenobiotic carboxylic acids. Palmitoyl-CoA formation exhibited biphasic kinetics indicative of two isoforms, a high affinity (Km1 2.3 microM) low capacity form and a low affinity (Km2 831 microM) high capacity form. These forms were differentially inhibited by a range of xenobiotics. However, it would appear that the low affinity component may not contribute to any major extent to the formation of xenobiotic-CoAs in vivo. At a concentration of 1 mM, greater than 20% inhibition of the high affinity form was observed with the 2-arylpropionates, ibuprofen, naproxen, benoxaprofen, fenoprofen, indoprofen, ketoprofen, tiaprofenic acid and cicloprofen, the hypolipidaemics, nafenopin and ciprofibrate, and the herbicides, silvex and 2,4,5-trichlorophenoxyacetate. Valproic acid, clofibric acid, salicylic acid and 2,4-dichlorophenoxy-acetate were non-inhibitory at all concentrations studied (0.1-2.5 mM). Analysis of the type of inhibition established that only nafenopin (Ki 430 microM) and ciprofibrate (Ki 97 microM) were competitive inhibitors of palmitoyl-CoA formation suggesting that they bind at the active site and thus potentially function as alternative substrates for the peroxisomal ligase. Notably, clofibric acid which has previously been shown to form clofibroyl-CoA in peroxisomes did not interact with the palmitoyl-CoA ligase thereby suggesting that activation is mediated via an alternative peroxisomal CoA ligase. In addition, the xenobiotic inhibitors of the peroxisomal palmitoyl-CoA ligase differed from those previously reported for the equivalent microsomal enzyme suggesting that the organellar forms may be functionally distinct. This study establishes that numerous xenobiotic carboxylic acids interact with the peroxisomal palmitoyl-CoA ligase; however, it would appear that relatively few function as alternative substrates. The toxicological ramifications of peroxisomally mediated xenobiotic-CoA formation and the identification of other peroxisomal xenobiotic-CoA ligase(s) remain to be elucidated.


Assuntos
Ácidos Carboxílicos/farmacologia , Coenzima A Ligases/antagonistas & inibidores , Fígado/efeitos dos fármacos , Microcorpos/enzimologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Xenobióticos/farmacologia , Animais , Fenoprofeno/farmacologia , Cetoprofeno/farmacologia , Cinética , Fígado/enzimologia , Masculino , Naproxeno/farmacologia , Ratos
15.
Biochem Pharmacol ; 43(7): 1465-71, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1567471

RESUMO

Microsomal long chain fatty acid CoA ligase (EC 6.2.1.3) has been implicated in the formation of CoA thioesters of xenobiotics containing a carboxylic acid moiety. In this study we have demonstrated that the microsomal enzyme from rat liver exhibits biphasic kinetics for the formation of palmitoyl-CoA, i.e. there are high affinity low capacity Kmhigh, 1.6 microM, Vmaxhigh, 12.9 nmol/mg/min) and low affinity high capacity (Kmlow, 506 microM, Vmaxlow, 58.3 nmol/mg/min) components. Inhibition of the high affinity isoform was studied using the R and S enantiomers of ibuprofen, fenoprofen, ketoprofen and naproxen. The high affinity component of palmitoyl-CoA formation was competitively inhibited by R-fenoprofen (Ki 15.4 microM) while R-ibuprofen exhibited mixed inhibition kinetics. In contrast the R and S enantiomers of ketoprofen and naproxen were non-competitive inhibitors. This diversity of inhibition kinetics observed argues in favour of a binding site in addition to the catalytic site. A competitive interaction with the high affinity form correlated with literature evidence of enantiospecific chiral inversion and "hybrid" triglyceride formation for the R enantiomers of fenoprofen and ibuprofen. Paradoxically, R-ketoprofen which is extensively inverted in rats was a non-competitive inhibitor of palmitoyl-CoA formation by the high affinity isoform suggesting that it may not act as an alternate substrate. The results of this study clearly indicate that formation of R-2-arylpropionate-CoAs is not fully explained by interaction with the high affinity isoform of a microsomal long chain (palmitoyl) CoA ligase and therefore the involvement of other isoforms cannot be discounted.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Coenzima A Ligases/antagonistas & inibidores , Microssomos Hepáticos/enzimologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Animais , Ligação Competitiva , Fenoprofeno/farmacologia , Ibuprofeno/farmacologia , Cetoprofeno/farmacologia , Cinética , Masculino , Naproxeno/farmacologia , Palmitoil Coenzima A/biossíntese , Ratos , Ratos Endogâmicos , Estereoisomerismo , Especificidade por Substrato , Triglicerídeos/biossíntese
16.
Biochem Pharmacol ; 42(7): 1421-5, 1991 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-1930265

RESUMO

Recent studies have shown that acyl-glucuronide conjugates are chemically reactive electrophilic metabolites that can undergo transacylation reactions resulting in intra-molecular rearrangement, hydrolysis and covalent binding of aglycone to albumin both in vitro and in vivo. The hypolipidaemic agent clofibrate is eliminated almost entirely as clofibric acid glucuronide in humans and rats. The formation of clofibric acid-protein adducts was investigated in 14 patients receiving 0.5-2.0 g/day of clofibrate for hypercholesterolaemia, and in liver homogenates from 20 rats administered 280 mg/kg/day of clofibric acid for up to 21 days. Total clofibric acid concentrations in the patients ranged from 0 to 114 mg/L. Covalently bound clofibric acid-protein adducts were detected in all patients, even in one subject in whom there was no measurable plasma clofibric acid. Concentrations ranged from 2.2 to 53.4 ng/mg protein and, in eight patients receiving 1.0 g/day of clofibrate, were correlated (P less than 0.05) with renal function as assessed by creatinine clearance. Clofibric acid-protein adducts were also present in rat liver homogenates, and increased with increasing duration of treatment (P less than 0.0001), from a mean (SE) of 10.1 (0.7) to 32.3 (1.6) ng/mg protein. The covalent binding of drugs to tissue macromolecules has traditionally been associated with toxicity. Further research is required to elucidate the role of acyl-glucuronide conjugates in the formation of drug-protein adducts and their biological consequences.


Assuntos
Proteínas Sanguíneas/metabolismo , Ácido Clofíbrico/farmacocinética , Fígado/metabolismo , Proteínas/metabolismo , Animais , Ácido Clofíbrico/sangue , Ácido Clofíbrico/uso terapêutico , Creatinina/sangue , Glucuronatos/metabolismo , Glucuronatos/urina , Humanos , Hipercolesterolemia/tratamento farmacológico , Masculino , Ratos , Análise de Regressão
19.
Pharmacol Toxicol ; 63(5): 327-32, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3237593

RESUMO

Hepatic damage was induced in phenobarbitone pretreated male Fischer 344 rats by the administration of 1% halothane in 14% oxygen for either 1 or 2 hours. Ethane production during the exposure period was not significantly different between the halothane and non-halothane exposed groups. Animals were sacrificed 1, 2, 6 and 24 hrs from commencement of anaesthesia and the hepatic microsomal fraction analyzed for diene conjugates, lipid hydroperoxides, total lipid content and fatty acid composition. Animals exposed to halothane and sacrificed at 2 and 24 hrs had significantly elevated levels of diene conjugates (P less than 0.05), while lipid hydroperoxide concentration and serum alanine aminotransferase increased in only those animals sacrificed at 24 hrs. Alterations in total lipid content and hepatic microsomal fatty acid composition were not observed in animals sacrificed after 1 and 2 hrs. A significant reduction in total lipid and arachidonic acid content occurred only in those animals sacrificed 24 hrs after exposure, however a concomitant increase in the saturated fatty acid fraction was not observed. It is proposed that alterations in fatty acid composition in vivo and evidence of lipid peroxidation occur as a result of cell death rather than an initiating event in halothane induced hepatic necrosis in rats.


Assuntos
Halotano/toxicidade , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/patologia , Necrose/induzido quimicamente , Alanina Transaminase/sangue , Animais , Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos , Fígado/efeitos dos fármacos , Masculino , Microssomos Hepáticos/metabolismo , Fenobarbital/toxicidade , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...