Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Biochem Pharmacol ; 50(11): 1933-41, 1995 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-8615875

RESUMO

The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) can be activated metabolically by cytochrome(s) P450 to DNA-damaging agents that result in the formation of tumors in various organs of several animal models. In the present study, 30-min incubations at 37 degrees containing 5 mg/mL pooled human placental microsomes, 36 nmol NNK (including 2 microCi [5-3H]NNK) and a 5 mM concentration of either NADH, NADPH, or both cofactors together resulted in the formation of 11.43 +/- 0.32, 35.40 +/- 4.64, and 44.05 +/- 1.66 pmol 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL)/mg protein/min (mean +/- SD, N = 3), respectively. Similar experiments using 7, 9, and 11 mM NADH, NADPH, and both cofactors together in equimolar concentrations yielded results that suggest that NADH- and NADPH-dependent reductions of NNK are catalyzed by different enzymes. Computer simulations for the production of NNAL based on various kinetic models corroborated the conclusion drawn from the empirically derived data. In human placental microsomes, the Km,app and Vmax,app for the formation of NNAL were 1021.9 +/- 251.5 microM and 4360.7 +/- 991.7 pmol/mg protein/min, respectively. Inhibition of cytochrome P450-dependent activities by carbon monoxide and dicumarol (100 and 200 microM) resulted in an average increase of NNAL production of 40 and 56%, respectively, suggesting that P450-dependent biotransformation of NNK is occurring in the absence of inhibitors. Similarly, polyclonal goat IgG against rabbit P450 reductase resulted in a 12% increase in the production of NNAL when compared with control values. Thirty micromolar rutin, ethacrynic acid, cibacron blue 3GA, and iodoacetic acid, known inhibitors of certain human carbonyl reductase(s), incubated with placental microsomes containing an equimolar concentration of NNK, did not have a significant effect on the production of NNAL. These results establish that: (1) cytochromes P450 are likely involved in the metabolism of NNK by human placental microsomes, (2) metabolism of NNK to NNAL by human placental microsomes is catalyzed by an NADPH-dependent carbonyl reductase(s) and an NADH-dependent carbonyl reductase(s), and (3) reduction of NNK to NNAL is catalyzed by a placental microsomal carbonyl reductase(s) not previously described.


Assuntos
Carcinógenos/metabolismo , Nitrosaminas/metabolismo , Placenta/metabolismo , Oxirredutases do Álcool/metabolismo , Simulação por Computador , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Cinética , Pulmão/metabolismo , Microssomos/metabolismo , Microssomos Hepáticos/metabolismo , NAD/metabolismo , NADP/metabolismo
2.
Biochem Pharmacol ; 48(6): 1273-80, 1994 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-7945421

RESUMO

A-esterase(s), an enzyme(s) that hydrolyzes certain organophosphate compounds, is located in mammals, primarily in serum and liver. Although considerable information is available regarding serum A-esterase(s), little is known about the hepatic form(s) of this enzyme. In the present study, hepatic A-esterase activity was quantified by measuring the EDTA-sensitive hydrolysis of the organophosphate paraoxon (O,O-diethyl-O-p-nitrophenyl phosphate). EDTA-insensitive hydrolysis was assumed to be the nonenzymatic phosphorylation of proteins with appropriate serine hydroxyl groups. Resuspension of mouse hepatic microsomes in 50 mM potassium phosphate buffer, pH 7.4, containing 100 microM calcium chloride, 0.25% sodium cholate, and 0.1% Triton N-101, resulted in the solubilization of A-esterase activity, as evidenced by the failure of activity to sediment after centrifugation at 100,000 g for 1 hr. Gel permeation chromatography followed by ion-exchange chromatography and nonspecific affinity chromatography resulted in a peak of A-esterase activity judged to be homogeneous by SDS-PAGE. A typical purification resulted in a 1531-fold increase in specific activity, with a recovery of 10%. SDS-PAGE with and without an acrylamide gradient indicated a molecular weight of 40,000 and 39,000 Da, respectively, while analyses of amino acid composition revealed similarities with human and rabbit serum paraoxonase. And finally, although this protein hydrolyzed both paraoxon and methyl paraoxon (O,O-dimethyl-O-p-nitrophenyl phosphate), it did not hydrolyze p-nitrophenyl acetate.


Assuntos
Esterases/isolamento & purificação , Microssomos Hepáticos/enzimologia , Aminoácidos/análise , Animais , Citosol/enzimologia , Esterases/química , Feminino , Masculino , Camundongos , Paraoxon/análogos & derivados , Paraoxon/metabolismo , Perfusão
3.
Biochem Pharmacol ; 50(11): 1925-32, 1995 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-8615874

RESUMO

Phosphorothioate insecticides such as parathion (O,O-diethyl-O-p-nitrophenyl phosphorothioate) undergo P450-dependent oxidative desulfuration, leading to both activation and detoxification of these compounds. Consequently, alterations in P450-dependent oxidative desulfuration may affect the acute toxicities of these insecticides. In the present study, pretreatment of mice with 15% ethanol in the drinking water for 6 days antagonized the acute toxicity of parathion, but not its toxic metabolite paraoxon (O,O-diethyl-O-p-nitrophenyl phosphate), suggesting that ethanol affected the oxidative desulfuration of this insecticide. The presence of ethanol within hepatic microsomal incubations did not alter the P450-dependent formation of paraoxon (activation) and p-nitrophenol (detoxification), although p-nitrophenol levels were increased in the presence of ethanol as a result of inhibition of its biotransformation to 4-nitrocatechol by CYP2E1. Ethanol exposure reduced hepatic pyruvate levels, but had no effect on levels of lactate, isocitrate, alpha-ketoglutarate, and malate. Calculation of cytosolic NAD+/NADH and cytosolic NADP+/NADPH redox ratios did not reveal any detectable difference in redox state between control and ethanol-treated mice. Since ethanol did not alter directly the P450-dependent activation or detoxification of parathion, and did not decrease NADPH levels, ethanol's antagonism of the acute toxicity of parathion may result from reduced availability of O2.


Assuntos
Etanol/administração & dosagem , Inseticidas/toxicidade , Fígado/metabolismo , Paration/toxicidade , Animais , Biotransformação/efeitos dos fármacos , Citocromo P-450 CYP2E1 , Sistema Enzimático do Citocromo P-450/metabolismo , Citosol/metabolismo , Etanol/sangue , Etanol/farmacologia , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Paraoxon/metabolismo , Paration/antagonistas & inibidores , Paration/química
4.
Environ Health Perspect ; 109 Suppl 1: 101-11, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11250810

RESUMO

We review pharmacokinetic and pharmacodynamic factors that should be considered in the design and interpretation of developmental neurotoxicity studies. Toxicologic effects on the developing nervous system depend on the delivered dose, exposure duration, and developmental stage at which exposure occurred. Several pharmacokinetic processes (absorption, distribution, metabolism, and excretion) govern chemical disposition within the dam and the nervous system of the offspring. In addition, unique physical features such as the presence or absence of a placental barrier and the gradual development of the blood--brain barrier influence chemical disposition and thus modulate developmental neurotoxicity. Neonatal exposure may depend on maternal pharmacokinetic processes and transfer of the xenobiotic through the milk, although direct exposure may occur through other routes (e.g., inhalation). Measurement of the xenobiotic in milk and evaluation of biomarkers of exposure or effect following exposure can confirm or characterize neonatal exposure. Physiologically based pharmacokinetic and pharmacodynamic models that incorporate these and other determinants can estimate tissue dose and biologic response following in utero or neonatal exposure. These models can characterize dose--response relationships and improve extrapolation of results from animal studies to humans. In addition, pharmacologic data allow an experimenter to determine whether exposure to the test chemical is adequate, whether exposure occurs during critical periods of nervous system development, whether route and duration of exposure are appropriate, and whether developmental neurotoxicity can be differentiated from direct actions of the xenobiotic.


Assuntos
Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/crescimento & desenvolvimento , Xenobióticos/farmacologia , Xenobióticos/farmacocinética , Animais , Biomarcadores/análise , Relação Dose-Resposta a Droga , Humanos , Modelos Biológicos , Ratos , Projetos de Pesquisa , Medição de Risco , Testes de Toxicidade/métodos , Xenobióticos/efeitos adversos
5.
Toxicol Sci ; 58(1): 118-26, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11053548

RESUMO

The mechanism of acute toxicity of the organophosphorus insecticides has been known for many years to be inhibition of the critical enzyme acetylcholinesterase (EC 3.1.1.7), with the resulting excess acetylcholine accumulation leading to symptoms of cholinergic excess. The bimolecular inhibition rate constant k(i) has been used for decades to describe the inhibitory capacity of organophosphates toward acetylcholinesterase. In the current study, a new approach based on continuous systems modeling was used to determine the appk(i)s of paraoxon and methyl paraoxon towards mouse brain acetylcholinesterase over a wide range of oxon concentrations. These studies revealed that the bimolecular inhibition rate constants for paraoxon and methyl paraoxon appeared to change as a function of oxon concentrations. For example, the appk(i) found with a paraoxon concentration of 1000 nM was 0.16 nM-1h-1, whereas that for 0.1 nM paraoxon was 1.60 nM-1h-1, indicating that the efficiency of phosphorylation appeared to decrease as the paraoxon concentration increased. These data suggested that the current understanding of how these organophosphates interact with acetylcholinesterase is incomplete. Modeling studies using several different kinetic schemes, as well as studies using recombinant monomeric mouse brain acetylcholinesterase, suggested the existence of a second binding site in addition to the active site of the enzyme, to which paraoxon and methyl paraoxon bound, probably in a reversibly manner. Occupation of this site likely rendered more difficult the subsequent phosphorylation of the active site by other oxon molecules, probably by steric hindrance or allosteric modification of the active site. It cannot be ascertained from the current study whether the putative second binding site is identical to or shares common elements with the well-characterized propidium-specific peripheral binding site of acetylcholinesterase.


Assuntos
Acetilcolinesterase/metabolismo , Encéfalo/enzimologia , Inseticidas/metabolismo , Paraoxon/análogos & derivados , Paraoxon/metabolismo , Animais , Sítios de Ligação , Simulação por Computador , Interações Medicamentosas , Masculino , Camundongos , Modelos Biológicos
6.
Toxicol Sci ; 63(2): 208-13, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11568364

RESUMO

Inhibition of the critical enzyme acetylcholinesterase (E.C. 3.1.1.7) with subsequent cholinergic crisis is the mechanism of acute toxicity of the organophosphorus insecticides (B. E. Mileson et al., 1998, Toxicol. Sci.41, 8-20). Consequently, measurement of acetylcholinesterase activity is important for evaluating the mammalian toxicity of this commonly used class of insecticides. While mammalian acetylcholinesterase activity has often been determined in tissue homogenates in the presence of the nondenaturing detergent Triton X-100 at a concentration of 1%, the potential actions of this detergent on the activity of this critical enzyme are not understood. In the current study, homogenization of rat brain in buffer containing 1% Triton X-100 slightly elevated the (app)V(max) for hydrolysis of acetylthiocholine, without affecting the (app)K(m) or the (app)K(ss). However, the presence of both 1% Triton X-100 and paraoxon (at concentrations of 5 nM-100 nM) resulted in complex kinetic interactions with acetylcholinesterase, as evidenced by a curvilinear secondary plot for determination of the (app)k(i). These results suggest that measurement of acetylcholinesterase activity in the presence of up to 1% Triton X-100, but in the absence of oxon, should pose no problems with regard to data interpretation, provided it is recognized that the detergent slightly elevates activity. However, measurement of acetylcholinesterase activity after enzyme was exposed simultaneously to Triton X-100 and oxon could be problematic. Caution is warranted when interpreting data where acetylcholinesterase activity was determined under such conditions since in the presence of 1% Triton X-100, the capacity of oxon to inhibit acetylcholinesterase might change as a function of oxon levels.


Assuntos
Acetilcolina/farmacologia , Acetilcolinesterase/metabolismo , Encéfalo/metabolismo , Detergentes/metabolismo , Inseticidas/metabolismo , Octoxinol/metabolismo , Paraoxon/metabolismo , Animais , Sítios de Ligação , Encéfalo/efeitos dos fármacos , Interações Medicamentosas , Masculino , Modelos Biológicos , Farmacocinética , Ratos , Ratos Sprague-Dawley
7.
Toxicol Sci ; 80(2): 239-48, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15141101

RESUMO

The primary mechanism of action for organophosphorus (OP) insecticides, like chlorpyrifos and parathion, is to inhibit acetylcholinesterase (AChE) by their oxygenated metabolites (oxons), due to the phosphorylation of the serine hydroxyl group located in the active site of the molecule. The rate of phosphorylation is described by the bimolecular inhibitory rate constant (k(i)), which has been used for quantification of OP inhibitory capacity. It has been proposed that a peripheral binding site exists on the AChE molecule, which, when occupied, reduces the capacity of additional oxon molecules to phosphorylate the active site. The aim of this study was to evaluate the interaction of chlorpyrifos oxon (CPO) and paraoxon (PO) with rat brain AChE to assess the dynamics of AChE inhibition and the potential role of a peripheral binding site. The k(i) values for AChE inhibition determined at oxon concentrations of 1-100 nM were 0.206 +/- 0.018 and 0.0216 nM(-1)h(-1) for CPO and PO, respectively. The spontaneous reactivation rates of the inhibited AChE for CPO and PO were 0.084-0.087 (two determinations) and 0.091 +/- 0.023 h(-1), respectively. In contrast, the k(i) values estimated at a low oxon concentration (1 pM) were approximately 1,000- and 10,000-fold higher than those determined at high CPO and PO concentrations, respectively. At low concentrations, the k(i) estimates were approximately similar for both CPO and PO (150-180 [two determinations] and 300 +/- 180 nM(-1)h(-1), respectively). This implies that, at low concentrations, both oxons exhibited similar inhibitory potency in contrast to the marked difference exhibited at higher concentrations. These results support the potential importance of a secondary peripheral binding site associated with AChE kinetics, particularly at low, environmentally relevant concentrations.


Assuntos
Clorpirifos/análogos & derivados , Clorpirifos/farmacologia , Inibidores da Colinesterase/farmacologia , Paraoxon/farmacologia , Acetilcolinesterase/química , Acetilcolinesterase/metabolismo , Algoritmos , Animais , Sítios de Ligação/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Clorpirifos/química , Inibidores da Colinesterase/química , Relação Dose-Resposta a Droga , Técnicas In Vitro , Cinética , Masculino , Paraoxon/química , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
8.
Toxicol Sci ; 41(1): 8-20, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9520337

RESUMO

The Food Quality Protection Act of 1996 (FQPA) requires the EPA to consider "available information concerning the cumulative effects of such residues and other substances that have a common mechanism of toxicity ... in establishing, modifying, leaving in effect, or revoking a tolerance for a pesticide chemical residue." This directive raises a number of scientific questions to be answered before the FQPA can be implemented. Among these questions is: What constitutes a common mechanism of toxicity? The ILSI Risk Science Institute (RSI) convened a group of experts to examine this and other scientific questions using the organophosphorus (OP) pesticides as the case study. OP pesticides share some characteristics attributed to compounds that act by a common mechanism, but produce a variety of clinical signs of toxicity not identical for all OP pesticides. The Working Group generated a testable hypothesis, anticholinesterase OP pesticides act by a common mechanism of toxicity, and generated alternative hypotheses that, if true, would cause rejection of the initial hypothesis and provide criteria for subgrouping OP compounds. Some of the alternative hypotheses were rejected outright and the rest were not supported by adequate data. The Working Group concluded that OP pesticides act by a common mechanism of toxicity if they inhibit acetylcholinesterase by phosphorylation and elicit any spectrum of cholinergic effects. An approach similar to that developed for OP pesticides could be used to determine if other classes or groups of pesticides that share structural and toxicological characteristics act by a common mechanism of toxicity or by distinct mechanisms.


Assuntos
Inseticidas/toxicidade , Animais , Inibidores da Colinesterase/toxicidade , Interações Medicamentosas , Humanos , Compostos Organofosforados/toxicidade , Medição de Risco
9.
Toxicology ; 51(2-3): 191-200, 1988 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2459806

RESUMO

Following single-pass perfusion of mouse livers in situ with the organophosphate pesticide chlorpyrifos, the cholinesterase inhibitor chlorpyrifos oxon could not be detected in effluent perfusate. Steady-state, with respect to chlorpyrifos, was achieved in 36-48 min, at which time the extraction ratio was 0.46. The hepatic disposition of chlorpyrifos was not affected by changes in perfusate flow rates (provided the flow rates maintained viable livers), but was altered by changes in the free fraction of chlorpyrifos within perfusate. However, under no conditions did chlorpyrifos oxon appear in effluent perfusate. Pretreatment of mice with phenobarbital enhanced the production of chlorpyrifos oxon from chlorpyrifos by mouse hepatic microsomes in vitro, but antagonized the acute toxicity of chlorpyrifos. Phenobarbital pretreatment increased the steady-state extraction ratio of chlorpyrifos to 0.94, but did not lead to the presence of chlorpyrifos oxon in effluent perfusate. Thus the enhanced hepatic detoxification of chlorpyrifos following phenobarbital pretreatment probably accounts for the antagonism of the acute toxicity afforded by phenobarbital pretreatment.


Assuntos
Clorpirifos/farmacocinética , Fígado/metabolismo , Animais , Biotransformação , Clorpirifos/análogos & derivados , Clorpirifos/metabolismo , Masculino , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Perfusão , Fenobarbital/farmacologia , Piridonas/metabolismo , Soroalbumina Bovina/farmacologia
10.
Toxicology ; 68(1): 1-9, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1714638

RESUMO

The present study was undertaken to characterize the metabolic activation of the organophosphorus insecticides chlorpyrifos [O,O-diethyl O-(3,5,6-trichloro-2-pyridyl) phosphorothionate] and fenitrothion [O,O-dimethyl O-(3-methyl-p-nitrophenyl) phosphorothionate] by intact rat liver. Single-pass perfusions of rat livers with chlorpyrifos or fenitrothion to steady state conditions resulted in the appearance of their corresponding oxygen analogs in effluent. In addition, detoxification of chlorpyrifos oxon [O,O-diethyl O-(3,5,6-trichloro-2-pyridyl) phosphate] or fenitrooxon [O,O-dimethyl O-(3-methyl-p-nitrophenyl) phosphate] by rat blood did not proceed at a rate rapid enough to prevent passage of at least some of these chemicals from liver to extrahepatic tissues, suggesting that hepatic biotransformation of chlorpyrifos and fenitrothion by rat liver results in their net activation. Although male rat livers produced more chlorpyrifos oxon and fenitrooxon from chlorpyrifos and fenitrothion, respectively, than did livers from female rats, the acute toxicities of chlorpyrifos and fenitrothion were greater in females than in males. Therefore, differences in hepatic activation of chlorpyrifos and fenitrothion in males and females cannot account for the sex differences in their acute toxicities in the rat. Finally, S-methyl glutathione and S-p-nitrophenyl glutathione were not detected in effluent or bile of livers perfused with fenitrothion, suggesting that glutathione-mediated biotransformation of this insecticide does not occur to any significant degree in intact liver.


Assuntos
Clorpirifos/farmacocinética , Fenitrotion/farmacocinética , Fígado/metabolismo , Animais , Biotransformação , Clorpirifos/análogos & derivados , Clorpirifos/metabolismo , Feminino , Fenitrotion/análogos & derivados , Fenitrotion/metabolismo , Inativação Metabólica , Masculino , Perfusão , Ratos , Ratos Endogâmicos
11.
Toxicology ; 128(3): 207-18, 1998 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-9750043

RESUMO

Parathion, like most organophosphorus insecticides currently in use, must undergo cytochrome P450(P450)-dependent activation in order to exert its acute mammalian toxicity (cholinergic crisis). Since P450 isoforms play such an important role in mediating the toxicity of parathion and related insecticides, factors which significantly alter P450 activities, such as exposure to certain xenobiotics, can also be expected to affect the toxicity of these potentially hazardous insecticides. Cimetidine is a H2-histamine antagonist that has been shown to inhibit several P450-isoforms. In addition, administration of cimetidine has been reported to result in clinically significant pharmacokinetic interactions with a wide variety of drugs. In the present study coexposure to cimetidine and parathion resulted in a moderate increase in the toxicity of this pesticide. However, coexposure to cimetidine and paraoxon did not alter the toxicity of the organophosphate, indicating that cimetidine likely affected P450-dependent formation of paraoxon from parathion. In vitro incubations of mouse hepatic microsomes demonstrated that, in addition to reducing the velocity of P450-dependent metabolism of parathion, cimetidine increased the proportion of paraoxon formed (activation). and decreased the proportion of p-nitrophenol formed (detoxification). Since parathion is not eliminated significantly by other routes in the mouse, the bulk of parathion in vivo was metabolized by P450 (although more slowly) in the presence of cimetidine, leading to a greater amount of paraoxon produced, and therefore greater toxicity. Incubations with individual P450 isoforms suggested that cimetidine could act by inhibition of P450 isoforms that detoxify parathion to a greater degree than cimetidine-resistant isoforms, and/or cimetidine could alter the proportions of detoxification versus activation of certain individual isoforms.


Assuntos
Cimetidina/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Antagonistas dos Receptores H2 da Histamina/farmacologia , Inseticidas/farmacocinética , Inseticidas/toxicidade , Paraoxon/metabolismo , Paration/farmacocinética , Paration/toxicidade , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Método de Monte Carlo
12.
Toxicology ; 36(2-3): 159-69, 1985 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-4049428

RESUMO

Single-pass perfusion in situ of mouse livers with the organophosphate paraoxon resulted in formation of p-nitrophenol (PNP), p-nitrophenyl sulfate (PNPS), and p-nitrophenyl-beta-D-glucuronide (PNPG). Following initiation of perfusion of paraoxon steady--state conditions were achieved in 15-25 min, at which time the extraction ratio was 0.55 (S.D. = 0.05). This suggests the capacity of mouse liver to biotransform paraoxon is not as great as previously reported. At all concentrations of paraoxon examined the amount of PNPS produced exceeded that of PNPG. However, as the concentration of paraoxon increased the relative proportion of PNP to PNPS and PNPG increased, indicating the capacity of liver to biotransform paraoxon exceeded the capacity to biotransform PNP. Single-pass perfusion in situ of mouse livers with PNP resulted in production of PNPS and PNPG. As with paraoxon, steady-state conditions were achieved in 15-25 min. The extraction ratio of PNP, as well as the metabolic profile, changed markedly with varying concentrations of PNP. At PNP reservoir concentrations of 4 microM or less the extraction ratio of PNP was 1, with all PNP metabolized to PNPS. As PNP concentrations increased (up to 75 microM) both unchanged PNP and PNPG appeared in the effluent. Thus the hepatic biotransformation of PNP was clearly dependent on substrate concentration.


Assuntos
Fígado/metabolismo , Nitrofenóis/metabolismo , Paraoxon/metabolismo , Animais , Biotransformação , Glucuronatos/metabolismo , Glutationa/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Nitrobenzenos/metabolismo
13.
Life Sci ; 56(2): 125-34, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7823759

RESUMO

Human serum A-esterase is a calcium-dependent enzyme that hydrolyzes the organophosphate paraoxon by an Ordered Uni Bi kinetic mechanism. Incubation of various concentrations of calcium chloride with human serum A-esterase resulted in corresponding changes in appk3 and appE for the reaction, while appk2 was unaffected. Carboxyglutamic acid (CAG) prevented calcium chloride from altering appk3, but not appE. Similarly CAG reduced the calcium-stimulated nonenzymatic hydrolysis of paraoxon, as well as the calcium-stimulated de-phosphorylation of chymotrypsin phosphorylated by paraoxon. These results suggest that calcium plays two roles in the hydrolysis of paraoxon by A-esterase. Firstly, calcium is required in order to maintain an active site. In this capacity calcium might participate directly in the catalytic reaction, or it might be required in order to maintain the appropriate confirmation of the active site. And secondly, free calcium (or calcium weakly associated with A-esterase) facilitates the removal of diethyl phosphate from A-esterase, probably by polarizing the P = O bond of the diethyl phosphate-A-esterase intermediate, thereby rendering phosphorus more susceptible to nucleophilic attack by hydroxide ions.


Assuntos
Cálcio/fisiologia , Esterases/sangue , Paraoxon/metabolismo , Adulto , Arildialquilfosfatase , Feminino , Humanos , Hidrólise , Cinética , Masculino
14.
Toxicol Lett ; 68(3): 275-84, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8516780

RESUMO

The present study was undertaken in an attempt to reconcile the seemingly conflicting observations that glutathione-dependent biotransformation of methyl parathion (O,O-dimethyl-O-(4-nitrophenyl)phosphorothionate) by hepatic supernatant or partially purified glutathione S-transferases occurs in vitro, but not to any significant degree in vivo in the mouse. While incubation of 20 microM methyl parathion with glutathione-fortified 100,000 x g hepatic supernatant resulted in biotransformation of this insecticide, addition of the carbon monoxide exposed microsomal fraction (without NADPH) to the supernatant abolished this metabolism. HPLC analyses of the distribution of methyl parathion between 100,000 x g supernatant and carbon monoxide-exposed microsomes revealed that little methyl parathion could be recovered in the 100,000 x g supernatant, and that the bulk of this insecticide was associated with the microsomal fraction. Increasing the concentration of methyl parathion to 1 mM resulted in a greater fraction of methyl parathion found in the supernatant compared to that with 20 microM, although the bulk of methyl parathion remained associated with the microsomal fraction. While this increase in the fraction of substrate located within the supernatant led to limited glutathione-dependent metabolism of methyl parathion, it must be emphasized that a liver concentration of 1 mM methyl parathion is far greater than that which could be achieved in vivo. In conclusion, the results of the present study support the hypothesis that in the mouse, glutathione-dependent metabolism of methyl parathion does not occur to a significant degree in vivo because of its limited access to the soluble glutathione S-transferases.


Assuntos
Glutationa/metabolismo , Fígado/metabolismo , Metil Paration/metabolismo , Animais , Monóxido de Carbono/farmacologia , Formaldeído/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , NADP/metabolismo
15.
Toxicol Lett ; 60(1): 27-37, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1539180

RESUMO

The acute toxicity of organothiophosphate insecticides like parathion results from their metabolic activation by cytochromes P450. The present study is directed towards the characterization of cytochrome-P450-dependent metabolism of parathion by various mouse brain regions. Intraperitoneal administration of [35S]parathion to mice led to covalently bound [35S]sulfur in various tissues, indicating their capacity to oxidatively desulfurate this insecticide. Liver contained the greatest amount of covalently bound sulfur, and brain the least. Among individual brain regions the olfactory bulb and hypothalamus possessed the highest levels of sulfur binding when expressed on a per milligram tissue basis. However, when expressed on a per brain region basis, sulfur binding was greatest within the cortex as a result of the large mass of this region, compared to the hypothalamus and olfactory bulb. Incubation of the 78,000 x g fraction of mouse brain with parathion resulted in formation of p-nitrophenol, although paraoxon could not be detected. However, given the current understanding of parathion metabolism by cytochromes P450, and given that paraoxon can rapidly disappear through phosphorylation of serine hydroxyl groups, it is reasonable to assume that at least some paraoxon was formed. Production of p-nitrophenol required NADPH and was inhibited by carbon monoxide. In vitro incubations of parathion with the 78,000 x g fraction of mouse brain indicated that the hypothalamus and olfactory bulb had the greatest capacity to produce p-nitrophenol. These results demonstrate that various mouse brain regions possess different capacities to metabolize parathion.


Assuntos
Encéfalo/metabolismo , Paration/metabolismo , Animais , Biotransformação , Sistema Enzimático do Citocromo P-450/fisiologia , Masculino , Camundongos , Nitrofenóis/metabolismo , Enxofre/metabolismo
16.
Toxicol Lett ; 17(1-2): 43-8, 1983 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6623508

RESUMO

Trimethyltin (TMT) levels were determined in various tissues of male mice at 1, 2, 4, 6, 10 and 16 h after administration (4.26 mg/kg; i.p.). Peak TMT levels in kidneys, liver, blood, lungs and testes were observed at 1 h following administration. Penetration into the brain, skeletal muscle and adipose tissue was also observed where maximum TMT levels were achieved 6-16 h following administration. 16 h post-treatment, the order of mean tissue concentrations, of the compound was: liver greater than testes greater than kidneys greater than lungs greater than brain greater than skeletal muscle greater than adipose tissue greater than blood. TMT was retained at peak levels in most tissues until, by 16 h, the animals exhibited tremors and convulsions followed by death. The mean concentration of TMT in the brain associated with delayed (central nervous system) (CNS) excitability at 16 h was 1.53 micrograms/g of wet tissue. These results indicate that TMT rapidly distributes and, although water-soluble, persists in tissues following an i.p. administration.


Assuntos
Compostos de Trialquitina/metabolismo , Compostos de Trimetilestanho/metabolismo , Tecido Adiposo/metabolismo , Animais , Encéfalo/metabolismo , Cinética , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Músculos/metabolismo , Testículo/metabolismo , Distribuição Tecidual , Compostos de Trimetilestanho/toxicidade
17.
Toxicol Lett ; 55(1): 77-83, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1998199

RESUMO

Depletion of hepatic glutathione in the mouse by pretreatment with diethyl maleate (DEM) is known to potentiate the acute toxicities of many dimethyl-substituted organothiophosphate insecticides. However, certain studies have raised doubts regarding the participation of glutathione in the detoxification of methyl parathion in the mouse, and hence the putative mechanism of action of DEM-induced potentiation of this insecticide. The present study evaluates the hypothesis that DEM potentiates the acute toxicities of methyl parathion, methyl paraoxon, and fenitrothion by a mechanism other than glutathione depletion. One hour following pretreatment of mice with DEM (0.75 ml/kg i.p.), glutathione was markedly depleted and the acute toxicities of methyl parathion, methyl paraoxon and fenitrothion were potentiated. Administration of glutathione monoethyl ester (20 mmol/kg p.o.) to DEM-pretreated mice attenuated DEM-depletion of hepatic glutathione, or maintained glutathione at or above control levels. However, glutathione monoethyl ester did not alter the DEM-induced potentiation of the lethality of these insecticides. Furthermore, administration of glutathione monoethyl ester to naive mice increased hepatic glutathione levels, but did not affect the percentage of animals succumbing to a challenge dose of methyl parathion, methyl paraoxon, or fenitrothion. These data indicate that DEM potentiates the toxicity of methyl parathion, methyl paraoxon or fenitrothion by a mechanism unrelated to hepatic glutathione content.


Assuntos
Fenitrotion/toxicidade , Glutationa/análogos & derivados , Fígado/efeitos dos fármacos , Metil Paration/toxicidade , Paraoxon/análogos & derivados , Animais , Peso Corporal/efeitos dos fármacos , Sinergismo Farmacológico , Glutationa/metabolismo , Glutationa/farmacologia , Fígado/metabolismo , Masculino , Maleatos/farmacologia , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Paraoxon/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA