Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Pharmacol ; 96(3): 288-95, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26032640

ABSTRACT

Several drug compounds have failed in clinical trials due to extensive biotransformation by aldehyde oxidase (AOX) (EC 1.2.3.1). One of the main reasons is the difficulty in scaling clearance for drugs metabolised by AOX, from preclinical species to human. Using methotrexate as a probe substrate, we evaluated AOX metabolism in liver cytosol from human and commonly used laboratory species namely guinea pig, monkey, rat and rabbit. We found that the metabolism of methotrexate in rabbit liver cytosol was several orders of magnitude higher than any of the other species tested. The results of protein quantitation revealed that the amount of AOX1 in human liver was similar to rabbit liver. To understand if the observed differences in activity were due to structural differences, we modelled rabbit AOX1 using the previously generated human AOX1 homology model. Molecular docking of methotrexate into the active site of the enzyme led to the identification of important residues that could potentially be involved in substrate binding and account for the observed differences. In order to study the impact of these residue changes on enzyme activity, we used site directed mutagenesis to construct mutant AOX1 cDNAs by substituting nucleotides of human AOX1 with relevant ones of rabbit AOX1. AOX1 mutant proteins were expressed in Escherichia coli. Differences in the kinetic properties of these mutants have been presented in this study.


Subject(s)
Aldehyde Oxidase/metabolism , Antimetabolites, Antineoplastic/metabolism , Liver/chemistry , Methotrexate/metabolism , Aldehyde Oxidase/chemistry , Amino Acid Sequence , Animals , Antimetabolites, Antineoplastic/chemistry , Catalytic Domain , Guinea Pigs , Humans , Kinetics , Liver/enzymology , Macaca mulatta , Methotrexate/chemistry , Molecular Docking Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidation-Reduction , Rabbits , Rats , Rats, Sprague-Dawley , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Structural Homology, Protein , Substrate Specificity
2.
Xenobiotica ; 45(6): 495-502, 2015.
Article in English | MEDLINE | ID: mdl-25539458

ABSTRACT

1. Cattle are an important component of the human food chain. Drugs used either legally or illegally in cattle may therefore enter the food chain and it is thus important to understand pathways of drug metabolism in this species, including sulfation catalyzed by the sulfotransferases (SULTs). 2. In this study, we have analyzed the sulfation of 4-nitrophenol and other compounds in male and female bovine liver and characterized recombinant bovine SULT isoforms 1A1 and 1B1 expressed in Escherichia coli. 3. We found that, in contrast to most other mammalian species, the major phenol sulfotransferase SULT1A1 is not expressed in bovine liver. Rather SULT1B1 seems to be a major form in both male and female bovine liver. 4. We also identified kinetic differences between bovine and human SULT1A1 and, using the human SULT1A1 crystal structure, identified two amino acid positions in the active site of bovine SULT1A1 (Ile89Val and Phe247Val) that may be responsible for these differences.


Subject(s)
Liver/enzymology , Sulfotransferases/chemistry , Sulfotransferases/metabolism , Animals , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Cattle , Crystallography, X-Ray , Female , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Nitrophenols/pharmacokinetics , Nitrophenols/pharmacology , Sulfotransferases/genetics
3.
Drug Metab Dispos ; 42(8): 1334-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24824603

ABSTRACT

Anticancer agent 6-mercaptopurine (6MP) has been in use since 1953 for the treatment of childhood acute lymphoblastic leukemia (ALL) and inflammatory bowel disease. Despite being available for 60 years, several aspects of 6MP drug metabolism and pharmacokinetics in humans are unknown. Molybdoflavoenzymes such as aldehyde oxidase (AO) and xanthine oxidase (XO) have previously been implicated in the metabolism of this drug. In this study, we investigated the in vitro metabolism of 6MP to 6-thiouric acid (6TUA) in pooled human liver cytosol. We discovered that 6MP is metabolized to 6TUA through sequential metabolism via the 6-thioxanthine (6TX) intermediate. The role of human AO and XO in the metabolism of 6MP was established using the specific inhibitors raloxifene and febuxostat. Both AO and XO were involved in the metabolism of the 6TX intermediate, whereas only XO was responsible for the conversion of 6TX to 6TUA. These findings were further confirmed using purified human AO and Escherichia coli lysate containing expressed recombinant human XO. Xanthine dehydrogenase (XDH), which belongs to the family of xanthine oxidoreductases and preferentially reduces nicotinamide adenine dinucleotide (NAD(+)), was shown to contribute to the overall production of the 6TX intermediate as well as the final product 6TUA in the presence of NAD(+) in human liver cytosol. In conclusion, we present evidence that three enzymes, AO, XO, and XDH, contribute to the production of 6TX intermediate, whereas only XO and XDH are involved in the conversion of 6TX to 6TUA in pooled HLC.


Subject(s)
Aldehyde Oxidase/metabolism , Liver/enzymology , Liver/metabolism , Mercaptopurine/metabolism , Metabolic Detoxication, Phase I/physiology , Xanthine Dehydrogenase/metabolism , Xanthine Oxidase/metabolism , Adult , Aged , Cytosol/enzymology , Cytosol/metabolism , Escherichia coli/metabolism , Female , Humans , Male , Middle Aged , Recombinant Proteins/metabolism , Uric Acid/analogs & derivatives , Uric Acid/metabolism , Young Adult
4.
Drug Metab Dispos ; 42(4): 695-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24430612

ABSTRACT

When investigating the potential for xanthine oxidase (XO)-mediated metabolism of a new chemical entity in vitro, selective chemical inhibition experiments are typically used. Most commonly, these inhibition experiments are performed using the inhibitor allopurinol (AP) and commercially prepared human liver cytosol (HLC) as the enzyme source. For reasons detailed herein, it is also a common practice to perfuse livers with solutions containing AP prior to liver harvest. The exposure to AP in HLC preparations could obviously pose a problem for measuring in vitro XO activity. To investigate this potential problem, an HPLC-MS/MS assay was developed to determine whether AP and its primary metabolite, oxypurinol, are retained within the cytosol for livers that were treated with AP during liver harvest. Differences in enzymatic activity for XO and aldehyde oxidase (AO) in human cytosol that can be ascribed to AP exposure were also evaluated. The results confirmed the presence of residual AP (some) and oxypurinol (all) human liver cytosol preparations that had been perfused with an AP-containing solution. In every case where oxypurinol was detected, XO activity was not observed. In contrast, the presence of AP and oxypurinol did not appear to have an impact on AO activity. Pooled HLC that was purchased from a commercial source also contained residual oxypurinol and did not show any XO activity. In the future, it is recommended that each HLC batch is screened for oxypurinol and/or XO activity prior to testing for XO-mediated metabolism of a new chemical entity.


Subject(s)
Allopurinol/pharmacology , Cytosol/enzymology , Enzyme Inhibitors/pharmacology , Liver/enzymology , Oxypurinol/pharmacology , Xanthine Oxidase/metabolism , Aldehyde Oxidase/metabolism , Allopurinol/analysis , Allopurinol/metabolism , Chromatography, High Pressure Liquid , Cytosol/drug effects , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism , Female , Humans , Limit of Detection , Liver/drug effects , Male , Oxypurinol/analysis , Oxypurinol/metabolism , Perfusion , Tandem Mass Spectrometry , Tissue Culture Techniques/methods , Xanthine Oxidase/antagonists & inhibitors
5.
Drug Metab Dispos ; 41(10): 1852-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23918666

ABSTRACT

Aldehyde oxidase (AOX) is a cytosolic enzyme expressed across a wide range of species, including guinea pig and rhesus monkey. These species are believed to be the best preclinical models for studying human AOX-mediated metabolism. We compared AOX activity in rhesus monkeys, guinea pigs, and humans using phthalazine and N-[2-(dimethylamino)ethyl]acridone-4-carboxamide (DACA) as substrates and raloxifene as an inhibitor. Michaelis-Menten kinetics was observed for phthalazine oxidation in rhesus monkey, guinea pig, and human liver cytosol, whereas substrate inhibition was seen with DACA oxidase activity in all three livers. Raloxifene inhibited phthalazine and DACA oxidase activity uncompetitively in guinea pig, whereas mixed-mode inhibition was seen in rhesus monkey. Our analysis of the primary sequence alignment of rhesus monkey, guinea pig, and human aldehyde oxidase isoform 1 (AOX1) along with homology modeling has led to the identification of several amino acid residue differences within the active site and substrate entrance channel of AOX1. We speculate that some of these residues might be responsible for the differences observed in activity. Overall, our data indicate that rhesus monkeys and guinea pigs would overestimate intrinsic clearance in humans and would be unsuitable to use as animal models. Our study also showed that AOX metabolism in species is substrate-dependent and no single animal model can be reliably used to predict every drug response in humans.


Subject(s)
Aldehyde Oxidase/metabolism , Cytosol/enzymology , Cytosol/metabolism , Liver/enzymology , Liver/metabolism , Animals , Guinea Pigs , Humans , Kinetics , Macaca mulatta/metabolism , Male , Oxidation-Reduction , Phthalazines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...