Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Toxicol Appl Pharmacol ; 288(1): 114-20, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26212258

ABSTRACT

Parathion, a widely used organophosphate insecticide, is considered a high priority chemical threat. Parathion toxicity is dependent on its metabolism by the cytochrome P450 system to paraoxon (diethyl 4-nitrophenyl phosphate), a cytotoxic metabolite. As an effective inhibitor of cholinesterases, paraoxon causes the accumulation of acetylcholine in synapses and overstimulation of nicotinic and muscarinic cholinergic receptors, leading to characteristic signs of organophosphate poisoning. Inhibition of parathion metabolism to paraoxon represents a potential approach to counter parathion toxicity. Herein, we demonstrate that menadione (methyl-1,4-naphthoquinone, vitamin K3) is a potent inhibitor of cytochrome P450-mediated metabolism of parathion. Menadione is active in redox cycling, a reaction mediated by NADPH-cytochrome P450 reductase that preferentially uses electrons from NADPH at the expense of their supply to the P450s. Using human recombinant CYP 1A2, 2B6, 3A4 and human liver microsomes, menadione was found to inhibit the formation of paraoxon from parathion. Administration of menadione bisulfite (40mg/kg, ip) to rats also reduced parathion-induced inhibition of brain cholinesterase activity, as well as parathion-induced tremors and the progression of other signs and symptoms of parathion poisoning. These data suggest that redox cycling compounds, such as menadione, have the potential to effectively mitigate the toxicity of organophosphorus pesticides including parathion which require cytochrome P450-mediated activation.


Subject(s)
Cholinesterase Inhibitors/toxicity , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Insecticides/toxicity , Liver/drug effects , Organophosphate Poisoning/prevention & control , Parathion/toxicity , Vitamin K 3/pharmacology , Acetylcholinesterase/metabolism , Activation, Metabolic , Animals , Cholinesterase Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Female , GPI-Linked Proteins/metabolism , Humans , Insecticides/metabolism , Liver/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , NADP/metabolism , Organophosphate Poisoning/enzymology , Organophosphate Poisoning/etiology , Oxidation-Reduction , Paraoxon/metabolism , Paraoxon/toxicity , Parathion/metabolism , Rats, Long-Evans , Recombinant Proteins/metabolism , Time Factors , Vitamin K 3/metabolism
2.
J Biochem Mol Toxicol ; 29(12): 545-51, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26179144

ABSTRACT

Nuclear receptors and transcription factors regulate the mRNA expression of many drug metabolizing enzymes, including the carboxylesterases (Ces). However, there are few data regarding whether these changes in mRNA expression result in alteration of protein levels or activity. In the present study, we sought to determine the isoform-specific regulation of hepatic Ces mRNA expression and activity following the administration of pharmacological activators of the constitutive androstane receptor (CAR), pregnane X receptor (PXR), and nuclear factor E2-related protein (Nrf2) to mice. The CAR activator 1,4-bis-[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP) and PXR ligand pregnenolone-16a-carbonitrile (PCN) increased Ces mRNA expression of various Ces2 isoforms, whereas the Nrf2 activator butylated hydroxyanisole primarily reduced Ces3a mRNA expression and induced Ces1g mRNA. TCPOBOP and PCN increased Ces2 hydrolytic activity in an isoform-specific manner. Taken together, these data demonstrate that activation of CAR, PXR, and Nrf2 regulates not only Ces mRNA expression, but also isoform-specific activity.


Subject(s)
Carboxylesterase/genetics , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Trans-Activators/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Pregnenolone Carbonitrile/pharmacology , Pyridines/pharmacology , RNA, Messenger/genetics
3.
Toxicology ; 416: 15-22, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30685356

ABSTRACT

Hepatic carboxylesterases (Ces) catalyze the metabolism of drugs, environmental toxicants, and endogenous lipids and are known to be regulated by multiple nuclear receptors. Perfluorooctanoic acid (PFOA) is a synthetic fluorochemical that has been associated with dyslipidemia in exposed populations. In liver, PFOA can activate nuclear receptors such as PPARα, and alter the metabolism and excretion of chemicals. Here, we sought to test the ability of PFOA to modulate Ces expression and activity in the presence and absence of the PPARα receptor. For this purpose, male C57BL/6 NCrl mice were administered PFOA (1 or 3 mg/kg, po, 7 days) and livers collected for assessment of Ces expression and activity. PFOA increased Ces1 and 2 protein and activity. Notably, PFOA increased Ces1d, 1e, 1f, 1 g, 2c, and 2e mRNAs between 1.5- and 2.5-fold, while it decreased Ces1c and 2b. Activation of PPARα by PFOA was confirmed by up-regulation of Cyp4a14 mRNA. In a separate study of PFOA-treated wild-type (WT) and PPARα-null mice, induction of Ces 1e and 1f mRNA and in turn, Ces1 protein, was PPARα-dependent. Interestingly, in PPARα-null mice, Ces1c, 1d, 1 g, 2a, 2b, and 2e mRNAs and Ces2 protein were up-regulated by PFOA which contributed to sustained up-regulation of Ces activity, although to a lower extent than observed in WT mice. Activation of the CAR and PXR receptors likely accounted for up-regulation of select Ces1 and 2 subtypes in PPARα-null mice. In conclusion, the environmental contaminant PFOA modulates the expression and function of hepatic Ces enzymes, in part through PPARα.


Subject(s)
Caprylates/toxicity , Carboxylesterase/metabolism , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Liver/drug effects , PPAR alpha/deficiency , Animals , Carboxylesterase/genetics , Cytochrome P450 Family 4/genetics , Cytochrome P450 Family 4/metabolism , Gene Expression Regulation, Enzymologic , Liver/enzymology , Male , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/agonists , PPAR alpha/genetics
4.
Ann N Y Acad Sci ; 1378(1): 80-86, 2016 08.
Article in English | MEDLINE | ID: mdl-27441453

ABSTRACT

Accidental or intentional exposures to parathion, an organophosphorus (OP) pesticide, can cause severe poisoning in humans. Parathion toxicity is dependent on its metabolism by the cytochrome P450 (CYP) system to paraoxon (diethyl 4-nitrophenyl phosphate), a highly poisonous nerve agent and potent inhibitor of acetylcholinesterase. We have been investigating inhibitors of CYP-mediated bioactivation of OPs as a method of preventing or reversing progressive parathion toxicity. It is well recognized that NADPH-cytochrome P450 reductase, an enzyme required for the transfer of electrons to CYPs, mediates chemical redox cycling. In this process, the enzyme diverts electrons from CYPs to support chemical redox cycling, which results in inhibition of CYP-mediated biotransformation. Using menadione as the redox-cycling chemical, we discovered that this enzymatic reaction blocks metabolic activation of parathion in rat and human liver microsomes and in recombinant CYPs important to parathion metabolism, including CYP1A2, CYP2B6, and CYP3A4. Administration of menadione to rats reduces metabolism of parathion, as well as parathion-induced inhibition of brain cholinesterase activity. This resulted in inhibition of parathion neurotoxicity. Menadione has relatively low toxicity and is approved by the Food and Drug Administration for other indications. Its ability to block parathion metabolism makes it an attractive therapeutic candidate to mitigate parathion-induced neurotoxicity.


Subject(s)
Cholinesterase Inhibitors/toxicity , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Drug Delivery Systems/methods , Parathion/toxicity , Vitamin K 3/administration & dosage , Animals , Cholinesterase Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Humans , Insecticides/metabolism , Insecticides/toxicity , Organophosphate Poisoning/drug therapy , Organophosphate Poisoning/enzymology , Parathion/metabolism , Vitamin K 3/metabolism
5.
Appl Ergon ; 42(1): 71-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20615493

ABSTRACT

The amount of sleep obtained between shifts is influenced by numerous factors including the length of work and rest periods, the timing of the rest period relative to the endogenous circadian cycle and personal choices about the use of non-work time. The current study utilised a real-world live-in mining environment to examine the amount of sleep obtained when access to normal domestic, family and social activities was restricted. Participants were 29 mining operators (26 male, average age 37.4 ± 6.8 years) who recorded sleep, work and fatigue information and wore an activity monitor for a cycle of seven day shifts and seven night shifts (both 12h) followed by either seven or fourteen days off. During the two weeks of work participants lived on-site. Total sleep time was significantly less (p<0.01) while on-site on both day (6.1 ± 1.0 h) and night shifts (5.7 ± 1.5 h) than days off (7.4 ± 1.4 h). Further, night shift sleep was significantly shorter than day-shift sleep (p<0.01). Assessment of subjective fatigue ratings showed that the sleep associated with both days off and night shifts had a greater recovery value than sleep associated with day shifts (p<0.01). While on-site, participants obtained only 6h of sleep indicating that the absence of competing domestic, family and social activities did not convert to more sleep. Factors including shift start times and circadian influences appear to have been more important.


Subject(s)
Mining , Sleep , Work Schedule Tolerance , Adult , Circadian Rhythm , Fatigue/epidemiology , Female , Humans , Male , South Australia
SELECTION OF CITATIONS
SEARCH DETAIL