101
|
Hartmann T, Terao M, Garattini E, Teutloff C, Alfaro JF, Jones JP, Leimkühler S. The Impact of Single Nucleotide Polymorphisms on Human Aldehyde Oxidase. Drug Metab Dispos 2012. [DOI: 10.1124/dmd.111.043828 10.1124/dmd.112.043828err] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
102
|
Hartmann T, Terao M, Garattini E, Teutloff C, Alfaro JF, Jones JP, Leimkühler S. The impact of single nucleotide polymorphisms on human aldehyde oxidase. Drug Metab Dispos 2012; 40:856-64. [PMID: 22279051 DOI: 10.1124/dmd.111.043828] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aldehyde oxidase (AO) is a complex molybdo-flavoprotein that belongs to the xanthine oxidase family. AO is active as a homodimer, and each 150-kDa monomer binds two distinct [2Fe2S] clusters, FAD, and the molybdenum cofactor. AO has an important role in the metabolism of drugs based on its broad substrate specificity oxidizing aromatic aza-heterocycles, for example, N(1)-methylnicotinamide and N-methylphthalazinium, or aldehydes, such as benzaldehyde, retinal, and vanillin. Sequencing the 35 coding exons of the human AOX1 gene in a sample of 180 Italian individuals led to the identification of relatively frequent, synonymous, missense and nonsense single-nucleotide polymorphisms (SNPs). Human aldehyde oxidase (hAOX1) was purified after heterologous expression in Escherichia coli. The recombinant protein was obtained with a purity of 95% and a yield of 50 μg/l E. coli culture. Site-directed mutagenesis of the hAOX1 cDNA allowed the purification of protein variants bearing the amino acid changes R802C, R921H, N1135S, and H1297R, which correspond to some of the identified SNPs. The hAOX1 variants were purified and compared with the wild-type protein relative to activity, oligomerization state, and metal content. Our data show that the mutation of each amino acid residue has a variable impact on the ability of hAOX1 to metabolize selected substrates. Thus, the human population is characterized by the presence of functionally inactive hAOX1 allelic variants as well as variants encoding enzymes with different catalytic activities. Our results indicate that the presence of these allelic variants should be considered for the design of future drugs.
Collapse
Affiliation(s)
- Tobias Hartmann
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476 Potsdam, Germany
| | | | | | | | | | | | | |
Collapse
|
103
|
Xu L, Woodward C, Khan S, Prakash C. In Vitro Metabolism of BIIB021, an Inhibitor of Heat Shock Protein 90, in Liver Microsomes and Hepatocytes of Rats, Dogs, and Humans and Recombinant Human Cytochrome P450 Isoforms. Drug Metab Dispos 2012; 40:680-93. [DOI: 10.1124/dmd.111.043000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
104
|
Tayama Y, Sugihara K, Sanoh S, Miyake K, Kitamura S, Ohta S. Developmental Changes of Aldehyde Oxidase Activity and Protein Expression in Human Liver Cytosol. Drug Metab Pharmacokinet 2012; 27:543-7. [DOI: 10.2133/dmpk.dmpk-11-nt-124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
105
|
Akabane T, Gerst N, Naritomi Y, Masters JN, Tamura K. A Practical and Direct Comparison of Intrinsic Metabolic Clearance of Several Non-CYP Enzyme Substrates in Freshly Isolated and Cryopreserved Hepatocytes. Drug Metab Pharmacokinet 2012; 27:181-91. [DOI: 10.2133/dmpk.dmpk-11-rg-097] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
106
|
Sharma R, Strelevitz TJ, Gao H, Clark AJ, Schildknegt K, Obach RS, Ripp SL, Spracklin DK, Tremaine LM, Vaz ADN. Deuterium Isotope Effects on Drug Pharmacokinetics. I. System-Dependent Effects of Specific Deuteration with Aldehyde Oxidase Cleared Drugs. Drug Metab Dispos 2011; 40:625-34. [DOI: 10.1124/dmd.111.042770] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
107
|
Hutzler JM, Yang YS, Albaugh D, Fullenwider CL, Schmenk J, Fisher MB. Characterization of aldehyde oxidase enzyme activity in cryopreserved human hepatocytes. Drug Metab Dispos 2011; 40:267-75. [PMID: 22031625 DOI: 10.1124/dmd.111.042861] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Substrates of aldehyde oxidase (AO), for which human clinical pharmacokinetics are reported, were selected and evaluated in pooled mixed-gender cryopreserved human hepatocytes in an effort to quantitatively characterize AO activity. Estimated hepatic clearance (Cl(h)) for BIBX1382, carbazeran, O⁶-benzylguanine, zaleplon, and XK-469 using cryopreserved hepatocytes was 18, 17, 12, <4.3, and <4.3 ml · min⁻¹ · kg⁻¹, respectively. The observed metabolic clearance in cryopreserved hepatocytes was confirmed to be a result of AO-mediated metabolism via two approaches. Metabolite identification after incubations in the presence of H₂¹⁸O confirmed that the predominant oxidative metabolite was generated by AO, as expected isotope patterns in mass spectra were observed after analysis by high-resolution mass spectrometry. Second, clearance values were efficiently attenuated upon coincubation with hydralazine, an inhibitor of AO. The low exposure after oral doses of BIBX1382 and carbazeran (∼5% F) would have been fairly well predicted using simple hepatic extraction (f(h)) values derived from cryopreserved hepatocytes. In addition, the estimated hepatic clearance value for O⁶-benzylguanine was within ∼80% of the observed total clearance in humans after intravenous administration (15 ml · min⁻¹ · kg⁻¹), indicating a reasonable level of quantitative activity from this in vitro system. However, a 3.5-fold underprediction of total clearance was observed for zaleplon, despite the 5-oxo metabolite being clearly observed. These data taken together suggest that the use of cryopreserved hepatocytes may be a practical approach for assessing AO-mediated metabolism in discovery and potentially useful for predicting hepatic clearance of AO substrates.
Collapse
Affiliation(s)
- J Matthew Hutzler
- Boehringer-Ingelheim Pharmaceuticals Inc., Translational Research (Drug Metabolism and Pharmacokinetics), 175 Briar Ridge Road, R&D 10578, Ridgefield, CT 06877, USA.
| | | | | | | | | | | |
Collapse
|
108
|
Sanoh S, Nozaki K, Murai H, Terashita S, Teramura T, Ohta S. Prediction of Human Metabolism of FK3453 by Aldehyde Oxidase Using Chimeric Mice Transplanted with Human or Rat Hepatocytes. Drug Metab Dispos 2011; 40:76-82. [DOI: 10.1124/dmd.111.041954] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
109
|
Havemeyer A, Lang J, Clement B. The fourth mammalian molybdenum enzyme mARC: current state of research. Drug Metab Rev 2011; 43:524-39. [DOI: 10.3109/03602532.2011.608682] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
110
|
Akabane T, Tanaka K, Irie M, Terashita S, Teramura T. Case report of extensive metabolism by aldehyde oxidase in humans: Pharmacokinetics and metabolite profile of FK3453 in rats, dogs, and humans. Xenobiotica 2011; 41:372-84. [DOI: 10.3109/00498254.2010.549970] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
111
|
Tayama Y, Sugihara K, Sanoh S, Miyake K, Morita S, Kitamura S, Ohta S. Effect of Tea Beverages on Aldehyde Oxidase Activity. Drug Metab Pharmacokinet 2011; 26:94-101. [DOI: 10.2133/dmpk.dmpk-10-nt-078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
112
|
Strolin Benedetti M. FAD-dependent enzymes involved in the metabolic oxidation of xenobiotics. ANNALES PHARMACEUTIQUES FRANÇAISES 2010; 69:45-52. [PMID: 21296217 DOI: 10.1016/j.pharma.2010.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/04/2010] [Accepted: 10/06/2010] [Indexed: 11/26/2022]
Abstract
Although the majority of oxidative metabolic reactions are mediated by the CYP superfamily of enzymes, non-CYP-mediated oxidative reactions can play an important role in the metabolism of xenobiotics. Among the major oxidative enzymes, other than CYPs, involved in the oxidative metabolism of drugs and other xenobiotics, the flavin-containing monooxygenases (FMOs), the molybdenum hydroxylases [aldehyde oxidase (AO) and xanthine oxidase (XO)] and the FAD-dependent amine oxidases [monoamine oxidases (MAOs) and polyamine oxidases (PAOs)] are discussed in this minireview. In a similar manner to CYPs, these oxidative enzymes can also produce therapeutically active metabolites and reactive/toxic metabolites, modulate the efficacy of therapeutically active drugs or contribute to detoxification. Many of them have been shown to be important in endobiotic metabolism (e.g. XO, MAOs), and, consequently, interactions between drugs and endogenous compounds might occur when they are involved in drug metabolism. In general, most non-CYP oxidative enzymes (e.g. FMOs, MAOs) appear to be noninducible or much less inducible than the CYP system. Some of these oxidative enzymes exhibit polymorphic expression, as do some CYPs (e.g. FMO3). It is possible that the contribution of non-CYP oxidative enzymes to the overall metabolism of xenobiotics is underestimated, as most investigations of drug metabolism have been performed using experimental conditions optimised for CYP activity, although in some cases the involvement of non-CYP oxidative enzymes in xenobiotic metabolism has been inferred from not sufficient experimental evidence.
Collapse
|
113
|
Kudo M, Sasaki T, Ishikawa M, Hirasawa N, Hiratsuka M. Functional characterization of genetic polymorphisms identified in the promoter region of the xanthine oxidase gene. Drug Metab Pharmacokinet 2010; 25:599-604. [PMID: 20930425 DOI: 10.2133/dmpk.dmpk-10-nt-054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Xanthine oxidase (XO) catalyzes the oxidation of endogenous and exogenous purines and pyrimidines. In the present study, we investigated polymorphisms in the promoter region of the XO gene. Sequence variations in the 5'-flanking region were screened using denaturing high-performance liquid chromatography (DHPLC) on DNA samples from 196 unrelated Japanese individuals. Thirteen polymorphisms were identified and 13 haplotypes were classified by haplotype analysis. The promoter activities of these polymorphisms were measured by luciferase assay in the human hepatoma cell lines HepG2 and Huh-7. Transcriptional activity was significantly lower in cell lines transfected with the reporter construct containing 5-kb upstream fragments with -1756T than in those with wild-type -1756C. Our results indicate that genetic variation in the promoter region of XO may determine interindividual differences in XO gene expression.
Collapse
Affiliation(s)
- Mutsumi Kudo
- Department of Clinical Pharmacotherapeutics, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | |
Collapse
|
114
|
Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde Oxidase: An Enzyme of Emerging Importance in Drug Discovery. J Med Chem 2010; 53:8441-60. [DOI: 10.1021/jm100888d] [Citation(s) in RCA: 282] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- David C. Pryde
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| | - Deepak Dalvie
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, 10628 Science Center Drive, La Jolla, California 92121
| | - Qiyue Hu
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, 10628 Science Center Drive, La Jolla, California 92121
| | - Peter Jones
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340
| | - Thien-Duc Tran
- WorldWide Medicinal Chemistry, Pfizer Global Research and Development, Sandwich, Kent, CT13 9NJ, England
| |
Collapse
|
115
|
Havemeyer A, Grünewald S, Wahl B, Bittner F, Mendel R, Erdélyi P, Fischer J, Clement B. Reduction of N-Hydroxy-sulfonamides, Including N-Hydroxy-valdecoxib, by the Molybdenum-Containing Enzyme mARC. Drug Metab Dispos 2010; 38:1917-21. [DOI: 10.1124/dmd.110.032813] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
116
|
Alteration in the activity of oxidative enzymes in the tissues of male Wistar albino rats exposed to cadmium. Int J Occup Med Environ Health 2010; 23:55-62. [PMID: 20442063 DOI: 10.2478/v10001-010-0002-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE The objective of the present study was to investigate the effect of cadmium (Cd) on the activities of some oxidative enzymes [viz Aldehyde oxidase, AO (E.C. 1.2.3.1); Xanthine oxidase, XO (E.C. 1.2.3.2); Sulphite oxidase, SO (E.C.1.8.3.1.); and Monoamine oxidase, MO (E.C. 1.4.3.4)] in the liver and kidney. MATERIALS AND METHODS Male Wistar albino rats were administered 1, 2 and 4 mg Cd(2+)/kg body weight for one and three months. The activities of the oxidative enzymes were subsequently analyzed in the liver and kidney after both periods of exposure. RESULTS There was a dose dependent increase in liver and kidney Cd concentration in the test rats as compared to control after both periods of treatment with the liver retaining higher concentration of Cd than the kidney for each of the exposure dose. The oxidative enzymes were decreased in a dose dependent manner in the liver and kidney after both periods of treatment. The percentage inhibition of these enzymes was less in the liver of rats treated with Cd for three months relative to the one month treated rats for each of the exposure dose. Conversely, the inhibition of the activities of these enzymes in the kidney of rats in all the treatment groups was more pronounced after three months relative to the trend in the one month treated rats. However, the activities of the oxidative enzymes were higher in the liver as compared to the kidney in all the treatment groups after both durations of Cd treatment. CONCLUSION Based on the results obtained, it can be concluded that the inhibition of the oxidative enzymes by Cd may disturb metabolism of bioactive endogenous substances, exogenous components of food and some xenobiotics.
Collapse
|
117
|
Lee K, Park SK, Kwon BM, Kim K, Yu HE, Ryu J, Oh SJ, Lee KS, Kang JS, Lee CW, Kwon MG, Kim HM. Transport and metabolism of the antitumour drug candidate 2'-benzoyloxycinnamaldehyde in Caco-2 cells. Xenobiotica 2010; 39:881-8. [PMID: 19925380 DOI: 10.3109/00498250903216000] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The transport and metabolism of the antitumour drug candidate 2'-benzoyloxycinnamaldehyde (BCA) was characterized in Caco-2 cells. BCA disappeared rapidly from the donor side without being transported to the receiver side during its absorptive transport across Caco-2 cells. Its metabolites 2'-hydroxycinnamaldehyde (HCA) and o-coumaric acid (OCA) were formed in both the donor and the receiver sides. HCA, in a separate study, also disappeared rapidly from the donor side, mostly being converted to its oxidative metabolite OCA during its absorptive transport across Caco-2 cells. OCA was transported rapidly in the absorptive direction across Caco-2 cells with a P(app) of 25.4 +/- 1.0 x 10(-6) cm s(-1) (mean +/- standard deviation (SD), n = 3). OCA was fully recovered from both the donor and the receiver side throughout the time-course of this study. Formation of HCA from BCA was inhibited almost completely by bis(p-nitrophenyl)phosphate (BNPP), a selective inhibitor of carboxylesterases (CES), and phenylmethylsulfonyl fluoride (PMSF), a broad specificity inhibitor of esterases in Caco-2 cells, suggesting that this hydrolytic biotransformation was likely mediated predominantly by CES. Conversion of HCA to OCA was inhibited significantly by isovanillin, a selective inhibitor of aldehyde oxidase (AO). Inhibitors for xanthine oxidase (XO) and aldehyde dehydrogenase (ALDH), which are known to be involved in the oxidation of aldehydes to carboxylic acids, did not have a significant effect on the biotransformation of HCA to OCA in Caco-2 cells. In summary, the present work demonstrates that BCA is hydrolysed rapidly to HCA, followed by subsequent oxidation to OCA, in Caco-2 cells. The results provide a mechanistic understanding of the poor absorption and low bioavailability of BCA after oral administration.
Collapse
Affiliation(s)
- K Lee
- Bio-Evaluation Center, KRIBB, Chungbuk, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Diamond S, Boer J, Maduskuie TP, Falahatpisheh N, Li Y, Yeleswaram S. Species-Specific Metabolism of SGX523 by Aldehyde Oxidase and the Toxicological Implications. Drug Metab Dispos 2010; 38:1277-85. [DOI: 10.1124/dmd.110.032375] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
119
|
Smith MA, Marinaki AM, Sanderson JD. Pharmacogenomics in the treatment of inflammatory bowel disease. Pharmacogenomics 2010; 11:421-37. [PMID: 20235796 DOI: 10.2217/pgs.10.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, the benefits of early aggressive treatment paradigms for inflammatory bowel disease have emerged. Symptomatic improvement is no longer considered adequate; instead, the aim of treatment has become mucosal healing and altered natural history. Nonetheless, we still fail to achieve these end points in a large number of our patients. There are many reasons why patients fail to respond or develop toxicity when exposed to drugs used for inflammatory bowel disease, but genetic variation is likely to account for a significant proportion of this. Some examples, notably thiopurine methyltransferase polymorphism in thiopurine treatment, are already established in clinical practice. We present a review of the expanding literature in this field, highlighting many interesting developments in pharmacogenomics applied to inflammatory bowel disease and, where possible, providing guidance on the translation of these developments into clinical practice.
Collapse
Affiliation(s)
- Melissa A Smith
- Department of Gastroenterology, 1st Floor, College House, St Thomas' Hospital, Lambeth Palace Road, London, SE1 7EH, UK
| | | | | |
Collapse
|
120
|
Abstract
Together with xanthine oxidase, aldehyde oxidase (AO) is a major member of a relatively small family of molybdenum hydroxylases. Both enzymes are homodimers with a subunit molecular weight of about 150 kDa and exhibit catalytic activity only as a dimer. An AO subunit contains a molybdopterin cofactor, an FAD and two different 2Fe-2S redox centers. The enzyme catalyzes oxidation of a wide range of endogenous and exogenous aldehydes and N-heterocyclic aromatic compounds. N-heterocycle-containing drugs such as methotrexate, 6-mercaptopurine, cinchona alkaloids and famciclovir are oxidized by this enzyme. Marked species differences have been well documented for the AO-catalyzed metabolism of drugs including methotrexate and famciclovir. In addition, a large rat strain variation has also been demonstrated in the oxidation activity of benzaldehyde and methotrexate. Marked differences in species, large differences in rat strains and individual differences in AO activities in some rat strains have been reported. However, little has been elucidated about any related molecular biological mechanisms. We examined the mechanism of individual variations and strain difference of rat AO using the technology of molecular biology. Our recent studies regarding the inter- and intra-difference of AO activities in rats are described.
Collapse
Affiliation(s)
- Kunio Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Sendai, Japan.
| |
Collapse
|
121
|
Zhang X, Liu HH, Weller P, Zheng M, Tao W, Wang J, Liao G, Monshouwer M, Peltz G. In silico and in vitro pharmacogenetics: aldehyde oxidase rapidly metabolizes a p38 kinase inhibitor. THE PHARMACOGENOMICS JOURNAL 2010; 11:15-24. [DOI: 10.1038/tpj.2010.8] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
122
|
Rashidi MR, Nazemiyeh H. Inhibitory effects of flavonoids on molybdenum hydroxylases activity. Expert Opin Drug Metab Toxicol 2010; 6:133-52. [DOI: 10.1517/17425250903426164] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
123
|
Dalvie D, Zhang C, Chen W, Smolarek T, Obach RS, Loi CM. Cross-Species Comparison of the Metabolism and Excretion of Zoniporide: Contribution of Aldehyde Oxidase to Interspecies Differences. Drug Metab Dispos 2009; 38:641-54. [DOI: 10.1124/dmd.109.030783] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
124
|
Fukiya K, Itoh K, Yamaguchi S, Kishiba A, Adachi M, Watanabe N, Tanaka Y. A Single Amino Acid Substitution Confers High Cinchonidine Oxidation Activity Comparable with That of Rabbit to Monkey Aldehyde Oxidase 1. Drug Metab Dispos 2009; 38:302-7. [DOI: 10.1124/dmd.109.030064] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
125
|
Alfaro JF, Joswig-Jones CA, Ouyang W, Nichols J, Crouch GJ, Jones JP. Purification and mechanism of human aldehyde oxidase expressed in Escherichia coli. Drug Metab Dispos 2009; 37:2393-8. [PMID: 19741035 DOI: 10.1124/dmd.109.029520] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human aldehyde oxidase 1 (AOX1) has been subcloned into a vector suitable for expression in Escherichia coli, and the protein has been expressed. The resulting protein is active, with sulfur being incorporated in the molybdopterin cofactor. Expression levels are modest, but 1 liter of cells supplies enough protein for both biochemical and kinetic characterization. Partial purification is achieved by nickel affinity chromatography through the addition of six histidines to the amino-terminal end of the protein. Kinetic analysis, including kinetic isotope effects and comparison with xanthine oxidase, reveal similar mechanisms, with some subtle differences. This expression system will allow for the interrogation of human aldehyde oxidase structure/function relationships by site-directed mutagenesis and provide protein for characterizing the role of AOX1 in drug metabolism.
Collapse
Affiliation(s)
- Joshua F Alfaro
- Department of Chemistry, Washington State University, Pullman, WA 99163, USA
| | | | | | | | | | | |
Collapse
|
126
|
Liu P, Liang S, Wang BJ, Guo RC. Construction of expression system of rabbit aldehyde oxidase cDNA for the clarification of species differences. Eur J Drug Metab Pharmacokinet 2009; 34:205-11. [DOI: 10.1007/bf03191175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
127
|
Atack JR, Maubach KA, Wafford KA, O'Connor D, Rodrigues AD, Evans DC, Tattersall FD, Chambers MS, MacLeod AM, Eng WS, Ryan C, Hostetler E, Sanabria SM, Gibson RE, Krause S, Burns HD, Hargreaves RJ, Agrawal NGB, McKernan RM, Murphy MG, Gingrich K, Dawson GR, Musson DG, Petty KJ. In Vitro and in Vivo Properties of 3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA Receptor α5 Subtype-Selective Inverse Agonist. J Pharmacol Exp Ther 2009; 331:470-84. [DOI: 10.1124/jpet.109.157636] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
128
|
Smith MA, Marinaki AM, Arenas M, Shobowale-Bakre M, Lewis CM, Ansari A, Duley J, Sanderson JD. Novel pharmacogenetic markers for treatment outcome in azathioprine-treated inflammatory bowel disease. Aliment Pharmacol Ther 2009; 30:375-84. [PMID: 19500084 DOI: 10.1111/j.1365-2036.2009.04057.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Azathioprine (AZA) pharmacogenetics are complex and much studied. Genetic polymorphism in TPMT is known to influence treatment outcome. Xanthine oxidase/dehydrogenase (XDH) and aldehyde oxidase (AO) compete with TPMT to inactivate AZA. AIM To assess whether genetic polymorphism in AOX1, XDH and MOCOS (the product of which activates the essential cofactor for AO and XDH) is associated with AZA treatment outcome in IBD. METHODS Real-time PCR was conducted for a panel of single nucleotide polymorphism (SNPs) in AOX1, XDH and MOCOS using TaqMan SNP genotyping assays in a prospective cohort of 192 patients receiving AZA for IBD. RESULTS Single nucleotide polymorphism AOX1 c.3404A > G (Asn1135Ser, rs55754655) predicted lack of AZA response (P = 0.035, OR 2.54, 95%CI 1.06-6.13) and when combined with TPMT activity, this information allowed stratification of a patient's chance of AZA response, ranging from 86% in patients where both markers were favourable to 33% where they were unfavourable (P < 0.0001). We also demonstrated a weak protective effect against adverse drug reactions (ADRs) from SNPs XDH c.837C > T (P = 0.048, OR 0.23, 95% CI 0.05-1.05) and MOCOS c.2107A > C, (P = 0.058 in recessive model, OR 0.64, 95%CI 0.36-1.15), which was stronger where they coincided (P = 0.019). CONCLUSION These findings have important implications for clinical practice and our understanding of AZA metabolism.
Collapse
Affiliation(s)
- M A Smith
- Department of Gastroenterology, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Lee K, Kwon BM, Kim K, Ryu J, Oh SJ, Lee KS, Kwon MG, Park SK, Kang JS, Lee CW, Kim HM. Plasma pharmacokinetics and metabolism of the antitumour drug candidate 2'-benzoyloxycinnamaldehyde in rats. Xenobiotica 2009; 39:255-65. [PMID: 19280524 DOI: 10.1080/00498250802650069] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The pharmacokinetics and metabolism of 2'-benzoyloxycinnamaldehyde (BCA) was characterized in male Sprague-Dawley rats as part of the preclinical evaluations for developing this compound as an antitumour agent. BCA was not detected in the plasma following either intravenous or oral dose, whereas its putative metabolites 2'-hydroxycinnamaldehyde (HCA) and o-coumaric acid were present at considerable levels. In separate pharmacokinetics studies, HCA exhibited a high systemic clearance and a large volume of distribution, whereas both pharmacokinetic parameters were much lower for o-coumaric acid. The terminal half-life of both metabolites was approximately 2 h. BCA was converted rapidly to HCA in rat serum, liver microsomes and cytosol in vitro; HCA was subsequently converted to o-coumaric acid in a quantitative manner only in the liver cytosol. In addition, the formation of o-coumaric acid was inhibited significantly by menadione, a specific inhibitor for aldehyde oxidase. Taken collectively, the results suggest that the rapid systemic clearance of HCA is likely due mainly to hepatic clearance occurring from aldehyde oxidase-catalysed biotransformation to o- coumaric acid. In conclusion, the present work demonstrates that the anticancer drug candidate BCA is highly likely to work as its active metabolite HCA in the body.
Collapse
Affiliation(s)
- Kiho Lee
- Bio-Evaluation Center, KRIBB, Chungbuk, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Itoh K, Asakawa T, Hoshino K, Adachi M, Fukiya K, Watanabe N, Tanaka Y. Functional analysis of aldehyde oxidase using expressed chimeric enzyme between monkey and rat. Biol Pharm Bull 2009; 32:31-5. [PMID: 19122276 DOI: 10.1248/bpb.32.31] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aldehyde oxidase (AO) is a homodimer with a subunit molecular mass of approximately 150 kDa. Each subunit consists of about 20 kDa 2Fe-2S cluster domain storing reducing equivalents, about 40 kDa flavine adenine dinucleotide (FAD) domain and about 85 kDa molybdenum cofactor (MoCo) domain containing a substrate binding site. In order to clarify the properties of each domain, especially substrate binding domain, chimeric cDNAs were constructed by mutual exchange of 2Fe-2S/FAD and MoCo domains between monkey and rat. Chimeric monkey/rat AO was referred to one with monkey type 2Fe-2S/FAD domains and a rat type MoCo domain. Rat/monkey AO was vice versa. AO-catalyzed 2-oxidation activities of (S)-RS-8359 were measured using the expressed enzyme in Escherichia coli. Substrate inhibition was seen in rat AO and chimeric monkey/rat AO, but not in monkey AO and chimeric rat/monkey AO, suggesting that the phenomenon might be dependent on the natures of MoCo domain of rat. A biphasic Eadie-Hofstee profile was observed in monkey AO and chimeric rat/monkey AO, but not rat AO and chimeric monkey/rat AO, indicating that the biphasic profile might be related to the properties of MoCo domain of monkey. Two-fold greater V(max) values were observed in monkey AO than in chimeric rat/monkey AO, and in chimeric monkey/rat AO than in rat AO, suggesting that monkey has the more effective electron transfer system than rat. Thus, the use of chimeric enzymes revealed that 2Fe-2S/FAD and MoCo domains affect the velocity and the quantitative profiles of AO-catalyzed (S)-RS-8359 2-oxidation, respectively.
Collapse
Affiliation(s)
- Kunio Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
The molecular genetics of nicotine metabolism involves multiple polymorphic catalytic enzymes. Variation in metabolic pathways results in nicotine disposition kinetics that differ between individuals and ethnic groups. Twin studies indicate that a large part of this variance is genetic in origin, although environmental influences also contribute. The primary aim of this chapter is to review the current knowledge regarding the genetic variability in the enzymes that metabolize nicotine in humans. The focus is on describing the genetic polymorphisms that exist in cytochromes P450 (CYPs), aldehyde oxidase 1 (AOX1), UDP-glucuronosyltransferases (UGTs), and flavin-containing monooxygenase 3 (FMO3). Genetic studies have demonstrated that polymorphisms in CYP2A6, the primary enzyme responsible for nicotine breakdown, make a sizable contribution to the wide range of nicotine metabolic capacity observed in humans. Thus, special attention will be given to CYP2A6, because slower nicotine metabolism requires less frequent self-administration, and accordingly influences smoking behaviors. In addition, the molecular genetics of nicotine metabolism in nonhuman primates, mice, and rats will be reviewed briefly.
Collapse
Affiliation(s)
- Jill C Mwenifumbo
- Centre for Addiction & Mental Health and Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
132
|
Knutson CG, Rubinson EH, Akingbade D, Anderson CS, Stec DF, Petrova KV, Kozekov ID, Guengerich FP, Rizzo CJ, Marnett LJ. Oxidation and glycolytic cleavage of etheno and propano DNA base adducts. Biochemistry 2009; 48:800-9. [PMID: 19132922 PMCID: PMC2975463 DOI: 10.1021/bi801654j] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Non-invasive strategies for the analysis of endogenous DNA damage are of interest for the purpose of monitoring genomic exposure to biologically produced chemicals. We have focused our research on the biological processing of DNA adducts and how this may impact the observed products in biological matrixes. Preliminary research has revealed that pyrimidopurinone DNA adducts are subject to enzymatic oxidation in vitro and in vivo and that base adducts are better substrates for oxidation than the corresponding 2′-deoxynucleosides. We tested the possibility that structurally similar exocyclic base adducts may be good candidates for enzymatic oxidation in vitro. We investigated the in vitro oxidation of several endogenously occurring etheno adducts [1,N2-ε-guanine (1,N2-ε-Gua), N2,3-ε-Gua, heptanone-1,N2-ε-Gua, 1,N6-ε-adenine (1,N6-ε-Ade), and 3,N4-ε-cytosine (3,N4-ε-Cyt)] and their corresponding 2′-deoxynucleosides. Both 1,N2-ε-Gua and heptanone-1,N2-ε-Gua were substrates for enzymatic oxidation in rat liver cytosol; heteronuclear NMR experiments revealed that oxidation occurred on the imidazole ring of each substrate. In contrast, the partially or fully saturated pyrimidopurinone analogues [i.e., 5,6-dihydro-M1G and 1,N2-propanoguanine (PGua)] and their 2′-deoxynucleoside derivatives were not oxidized. The 2′-deoxynucleoside adducts, 1,N2-ε-dG and 1,N6-ε-dA, underwent glycolytic cleavage in rat liver cytosol. Together, these data suggest that multiple exocyclic adducts undergo oxidation and glycolytic cleavage in vitro in rat liver cytosol, in some instances in succession. These multiple pathways of biotransformation produce an array of products. Thus, the biotransformation of exocyclic adducts may lead to an additional class of biomarkers suitable for use in animal and human studies.
Collapse
Affiliation(s)
- Charles G Knutson
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
van Dijk AJ, Parvizi N, Taverne MAM, Fink-Gremmels J. Placental transfer and pharmacokinetics of allopurinol in late pregnant sows and their fetuses. J Vet Pharmacol Ther 2009; 31:489-95. [PMID: 19000269 DOI: 10.1111/j.1365-2885.2008.00976.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
At present no standard pharmacological intervention strategy is available to reduce these adverse effects of birth asphyxia. In the present study we aimed to evaluate placental transfer of allopurinol, an inhibitor of XOR. For this purpose, fetal catheterization of the jugular vein was conducted in five late pregnant sows (one fetus per sow). At 24-48 h after surgery, sows received allopurinol (15 mg/kg body weight; i.v.) and pharmacokinetics of allopurinol and its active metabolite oxypurinol were measured in both late pregnant sows and fetuses. Maternal and fetal blood samples were collected during and after allopurinol administration. Maternal C(max) values averaged 41.90 microg/mL (allopurinol) and 3.68 microg/mL (oxypurinol). Allopurinol crossed the placental barrier as shown by the average fetal C(max) values of 5.05 microg/mL at 1.47 h after allopurinol administration to the sow. In only one fetus low plasma oxypurinol concentrations were found. Incubations of subcellular hepatic fractions of sows and 24-h-old piglets confirmed that allopurinol could be metabolized into oxypurinol. In conclusion, we demonstrated that allopurinol can cross the placental barrier, a prerequisite for further studies evaluating the use of allopurinol as a neuroprotective agent to reduce the adverse effects following birth asphyxia in neonatal piglets.
Collapse
Affiliation(s)
- A J van Dijk
- Department of Veterinary Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
134
|
Itoh K, Adachi M, Sato J, Shouji K, Fukiya K, Fujii K, Tanaka Y. Effects of Selenium Deficiency on Aldehyde Oxidase 1 in Rats. Biol Pharm Bull 2009; 32:190-4. [DOI: 10.1248/bpb.32.190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kunio Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Mayuko Adachi
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Jun Sato
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Kanako Shouji
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Kensuke Fukiya
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Keiko Fujii
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Yorihisa Tanaka
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| |
Collapse
|
135
|
Jetter A, Kinzig M, Rodamer M, Tomalik-Scharte D, Sörgel F, Fuhr U. Phenotyping of N-acetyltransferase type 2 and xanthine oxidase with caffeine: when should urine samples be collected? Eur J Clin Pharmacol 2008; 65:411-7. [PMID: 19082994 DOI: 10.1007/s00228-008-0597-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 11/24/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Individual activities of N-acetyltransferase 2 (NAT2) and of xanthine oxidase (XO) can be assessed using ratios of urinary caffeine metabolites. We investigated how ratios changed over time and which urine collection interval would be the best for NAT2 and XO activity assessments. METHODS On two occasions separated by 14 days, 16 healthy male Caucasians collected urine before and 0-2, 2-4, 4-6, 6-8, 8-12, 12-16 and 16-24 h after a dose of 150 mg caffeine given in the framework of a phenotyping cocktail study. The metabolites 5-acetylamino-6-formylamino-3-methyluracil (AFMU), 5-acetylamino-6-amino-3-methyluracil (AAMU), 1-methylxanthine (1X), and 1-methylurate (1U) were quantified with LC-MS/MS. The molar ratio (AFMU + AAMU)/(1X + 1U + AFMU + AAMU) was used as a NAT2 metric, while the ratio 1U/(1X + 1U) served as XO metric. RESULTS The NAT2 ratios were stable in the intervals 4-24 h after caffeine dosing. Mean intra-individual coefficients of variation were 11-23% starting 4 h post-dose, while inter-individual variability reached 37-75%. The XO ratios increased gradually by 14% from the 2-4 to the 16-24 h interval. The mean intra- and inter-individual coefficients of variation of XO activity were 3-18 and 7-10% respectively. No significant differences between study occasions were observed. CONCLUSIONS Any sampling interval at least 4 h after caffeine dosing is suitable for NAT2 and XO activity assessments. XO activities can only be compared between volunteers and studies if the same urine collection schedule has been respected. The low intraindividual variability allows for sample sizes of 16 and 6 participants in crossover interaction studies of NAT2 and XO activity respectively.
Collapse
Affiliation(s)
- Alexander Jetter
- Department of Pharmacology, Clinical Pharmacology, University Hospital Cologne, Köln, Germany.
| | | | | | | | | | | |
Collapse
|
136
|
Pirouzpanah S, Hanaee J, Razavieh SV, Rashidi MR. Inhibitory effects of flavonoids on aldehyde oxidase activity. J Enzyme Inhib Med Chem 2008; 24:14-21. [DOI: 10.1080/14756360701841301] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Saeed Pirouzpanah
- School of Health and Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Hanaee
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed-Vali Razavieh
- School of Health and Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
137
|
Ohbuchi M, Miyata M, Nagai D, Shimada M, Yoshinari K, Yamazoe Y. Role of Enzymatic N-Hydroxylation and Reduction in Flutamide Metabolite-Induced Liver Toxicity. Drug Metab Dispos 2008; 37:97-105. [DOI: 10.1124/dmd.108.021964] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
138
|
Itoh K, Maruyama H, Adachi M, Hoshino K, Watanabe N, Tanaka Y. Lack of dimer formation ability in rat strains with low aldehyde oxidase activity. Xenobiotica 2008; 37:709-16. [PMID: 17620217 DOI: 10.1080/00498250701397713] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Aldehyde oxidase (AO) is a homodimer with a molecular weight of 300 kDa. To clarify the reasons for the well-known differences in rat strains, we set out to study the relationship between AO activity and the expression levels of its dimer. AO-catalyzed 2-oxidation activity of (S)-RS-8359 was measured in liver cytosols from ten rat strains. The expression levels of AO dimeric protein were evaluated by the native-PAGE/Western blot. Rat strains with low AO activity showed only a monomer, whereas strains with high activity overwhelmingly exhibited a dimer. Exceptionally, one strain in the high AO activity group displayed complex mixed expression patterns of low and high AO activity groups. However, there was a good relationship between AO activity and the expression levels of a dimer, but not of a monomer. The results suggest that rat strains with low AO activity lack the ability to produce a dimer necessary for catalytic activity, and AO differences in rat strains should be discussed in terms of the expression levels of the dimer itself.
Collapse
Affiliation(s)
- K Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Japan
| | | | | | | | | | | |
Collapse
|
139
|
Small-interference RNA-mediated knock-down of aldehyde oxidase 1 in 3T3-L1 cells impairs adipogenesis and adiponectin release. FEBS Lett 2008; 582:2965-72. [DOI: 10.1016/j.febslet.2008.07.034] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 07/07/2008] [Accepted: 07/20/2008] [Indexed: 12/14/2022]
|
140
|
Knutson CG, Skipper PL, Liberman RG, Tannenbaum SR, Marnett LJ. Monitoring in vivo metabolism and elimination of the endogenous DNA adduct, M1dG {3-(2-deoxy-beta-D-erythro-pentofuranosyl)pyrimido[1,2-alpha]purin-10(3H)-one}, by accelerator mass spectrometry. Chem Res Toxicol 2008; 21:1290-4. [PMID: 18461974 DOI: 10.1021/tx800049v] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Our laboratory is investigating the in vitro and in vivo metabolic processing of endogenously formed DNA adducts as a means of evaluating candidate urinary biomarkers. In particular, we have focused our studies on the metabolism and disposition of the peroxidation-derived pyrimidopurinone deoxyguanosine (dG) adduct, 3-(2-deoxy-beta-D-erythro-pentofuranosyl)pyrimido[1,2-R]purin-10(3H)-one (M1dG), and its principal metabolite, 6-oxo-M1dG. We now report the metabolic processing of M1dG at concentrations 4-8 orders of magnitude lower in concentration than previously analyzed, by the use of accelerator mass spectrometry analysis. Administration of 2.0 nCi/kg [14C]M1dG resulted in 49% of the 14C recovered in urine, whereas 51% was recovered in feces. In urine samples, approximately 40% of the 14C corresponded to the metabolite, 6-oxo-M1dG. Following iv administration of 0.5 and 54 pCi/kg [14C]M1dG, approximately 25% of the urinary recovery corresponded to the metabolite, 6-oxo-M1dG. Thus, upon administration of trace amounts of M1dG, a significant percentage of 6-oxo-M1dG was produced, suggesting that 6-oxo-M1dG maybe a useful urinary marker of exposure to endogenous oxidative damage.
Collapse
Affiliation(s)
- Charles G Knutson
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, USA
| | | | | | | | | |
Collapse
|
141
|
Abstract
OBJECTIVE Xanthine oxidase (XO) catalyzes the oxidation of endogenous and exogenous purines and pyrimidines. In this study, we speculated that individual variations in XO activity are caused by genetic variations in the XO gene. METHODS To investigate the genetic variations in XO in 96 Japanese participants, denaturing high-performance liquid chromatography was used. To assess the effects of these variations on enzymatic activity, wild-type XO and 21 types of variant XO--including those in the database and those just discovered--were transiently expressed in COS-7 cells. RESULTS Three nonsynonymous single nucleotide polymorphisms, including 514G>A (Gly172Arg), 3326A>C (Asp1109Thr), and 3662A>G (His1221Arg) were identified in Japanese participants. Functional characterization of 21 XO variants showed a deficiency in enzyme activity in two variants (Arg149Cys and Thr910Lys); low activity (intrinsic clearance, CLint: 22-69% compared with the wild-type) in six variants (Pro555Ser, Arg607Gln, Thr623Ile, Asn909Lys, Pro1150Arg, and Cys1318Tyr); and high activity (CLint: approximately two-fold higher than that in the wild-type) in two variants (Ile703Val and His1221Arg). CONCLUSION These results suggest that several single nucleotide polymorphisms in the XO gene are involved in individual variations in XO activity. In addition, such findings will be useful to identify xanthinuria patients.
Collapse
|
142
|
Kitamura S, Nitta K, Tayama Y, Tanoue C, Sugihara K, Inoue T, Horie T, Ohta S. Aldehyde Oxidase-Catalyzed Metabolism of N1-Methylnicotinamide in Vivo and in Vitro in Chimeric Mice with Humanized Liver. Drug Metab Dispos 2008; 36:1202-5. [DOI: 10.1124/dmd.107.019075] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
143
|
Adachi M, Itoh K, Masubuchi A, Watanabe N, Tanaka Y. Construction and expression of mutant cDNAs responsible for genetic polymorphism in aldehyde oxidase in Donryu strain rats. BMB Rep 2008; 40:1021-7. [PMID: 18047800 DOI: 10.5483/bmbrep.2007.40.6.1021] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We demonstrated the genetic polymorphism of aldehyde oxidase (AO) in Donryu strain rats: the ultrarapid metabolizer (UM) with nucleotide mutation of (377G, 2604C) coding for amino acid substitution of (110Gly, 852Val), extensive metabolizer (EM) with (377G/A, 2604C/T) coding for (110Gly/Ser, 852Val/Ala), and poor metabolizer (PM) with (377A, 2604T) coding for (110Ser, 852Ala), respectively. The results suggested that 377G > A and/or 2604C > T should be responsible for the genetic polymorphism. In this study, we constructed an E. coli expression system of four types of AO cDNA including Mut-1 with (377G, 2604T) and Mut-2 with (377A, 2604C) as well as naturally existing nucleotide sequences of UM and PM in order to clarify which one is responsible for the polymorphism. Mut-1 and Mut-2 showed almost the same high and low activity as that of the UM and PM groups, respectively. Thus, the expression study of mutant AO cDNA directly revealed that the nucleotide substitution of 377G > A, but not that of 2604C > T, will play a critical role in the genetic polymorphism of AO in Donryu strain rats. The reason amino acid substitution will cause genetic polymorphism in AO activity was discussed.
Collapse
Affiliation(s)
- Mayuko Adachi
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | |
Collapse
|
144
|
Asakawa T, Itoh K, Adachi M, Hoshino K, Watanabe N, Tanaka Y. Properties of 130 kDa Subunit of Monkey Aldehyde Oxidase. Biol Pharm Bull 2008; 31:380-5. [DOI: 10.1248/bpb.31.380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tasuku Asakawa
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Kunio Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Mayuko Adachi
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Kouichi Hoshino
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| | - Nobuaki Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi-Sankyo Co., Ltd
| | - Yorihisa Tanaka
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University
| |
Collapse
|
145
|
Adachi M, Itoh K, Abe H, Tanaka Y. Heredity mode of genetic polymorphism in aldehyde oxidase activity in Donryu strain rats. Xenobiotica 2008; 38:98-105. [PMID: 18098066 DOI: 10.1080/00498250701708513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Donryu strain rats show genetic polymorphisms in the aldehyde oxidase gene, resulting in the phenotypic expression of ultrarapid metabolizers with homozygous nucleotide sequences (337G, 2604C), extensive metabolizers with heterozygous nucleotide sequences (377G/A, 2604C/T), and poor metabolizers with homozygous nucleotide sequences (377A, 2604T). In the mating experiments the ratio of the number of ultrarapid metabolizers, extensive metabolizers, and poor metabolizers rats in the F1 generation from the heterozygous F0 extensive metabolizers male and female rats was roughly 0.6 : 1.5 : 1, and the ratio converged to approximately 1 : 2 : 1 in the F2 generation from the heterozygous F1 extensive metabolizers male and female rats. On the contrary, all the F2 generation from homozygous F1 ultrarapid metabolizers male and female rats or from homozygous F1 poor metabolizers male and female rats had the ultrarapid metabolizers or the poor metabolizers genotypes and phenotypes. The genotypes completely agreed with the phenotypes in all individuals of F0, F1, and F2 generations. The results indicate that the genetic polymorphism of aldehyde oxidase in Donryu strain rats obeys Mendelian heredity. The reason for a low ratio of the ultrarapid metabolizers rats in the commercially available Donryu strain rats - not more than several per cent - compared with the ratio expected from the Mendelian rule is unknown.
Collapse
Affiliation(s)
- M Adachi
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Aoba-ku, Sendai, Japan
| | | | | | | |
Collapse
|
146
|
Frampton JP, Shuler ML, Shain W, Hynd MR. Biomedical Technologies for in vitro Screening and Controlled Delivery of Neuroactive Compounds. Cent Nerv Syst Agents Med Chem 2008; 8:203-219. [PMID: 19079777 PMCID: PMC2600660 DOI: 10.2174/187152408785699613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell culture models can provide information pertaining to the effective dose, toxiciology, and kinetics, for a variety of neuroactive compounds. However, many in vitro models fail to adequately predict how such compounds will perform in a living organism. At the systems level, interactions between organs can dramatically affect the properties of a compound by alteration of its biological activity or by elimination of it from the body. At the tissue level, interaction between cell types can alter the transport properties of a particular compound, or can buffer its effects on target cells by uptake, processing, or changes in chemical signaling between cells. In any given tissue, cells exist in a three-dimensional environment bounded on all sides by other cells and components of the extracellular matrix, providing kinetics that are dramatically different from the kinetics in traditional two-dimensional cell culture systems. Cell culture analogs are currently being developed to better model the complex transport and processing that occur prior to drug uptake in the CNS, and to predict blood-brain barrier permeability. These approaches utilize microfluidics, hydrogel matrices, and a variety of cell types (including lung epithelial cells, hepatocytes, adipocytes, glial cells, and neurons) to more accurately model drug transport and biological activity. Similar strategies are also being used to control both the spatial and temporal release of therapeutic compounds for targeted treatment of CNS disease.
Collapse
Affiliation(s)
- John P Frampton
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | | | | | | |
Collapse
|
147
|
Pelletier J, Bozzolan F, Solvar M, François MC, Jacquin-Joly E, Maïbèche-Coisne M. Identification of candidate aldehyde oxidases from the silkworm Bombyx mori potentially involved in antennal pheromone degradation. Gene 2007; 404:31-40. [PMID: 17904312 DOI: 10.1016/j.gene.2007.08.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 08/21/2007] [Accepted: 08/21/2007] [Indexed: 10/22/2022]
Abstract
Signal inactivation is a crucial step in the dynamic of olfactory process and involves various Odorant-Degrading Enzymes. In the silkworm Bombyx mori, one of the best models for studying olfaction in insects, the involvement of an antennal-specific aldehyde oxidase in the degradation of the sex pheromone component bombykal has been demonstrated over the three past decades by biochemical studies. However, the corresponding enzyme has never been characterized at the molecular level. Bioinformatic screening of B. mori genome and molecular approaches have been used to isolate several candidate sequences of aldehyde oxidases. Two interesting antennal-expressed genes have been further characterized and their putative functions are discussed in regard to their respective expression pattern and to our knowledge on aldehyde oxidase properties. Interestingly, one gene appeared as specifically expressed in the antennae of B. mori and associated in males with the bombykal-sensitive sensilla, strongly suggesting that it could encode for the previously biochemically characterized enzyme.
Collapse
Affiliation(s)
- Julien Pelletier
- UMR-A 1272 Physiologie de l'Insecte: Signalisation et Communication, Université Pierre et Marie Curie-Paris 6, 7 Quai St-Bernard, Paris, France
| | | | | | | | | | | |
Collapse
|
148
|
Itoh K, Maruyama H, Adachi M, Hoshino K, Watanabe N, Tanaka Y. Lack of formation of aldehyde oxidase dimer possibly due to 377G>A nucleotide substitution. Drug Metab Dispos 2007; 35:1860-4. [PMID: 17639027 DOI: 10.1124/dmd.107.015503] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In addition to the many articles reporting on the marked differences in species and large differences in rat strains in response to aldehyde oxidase (AO), individual differences in some rat strains have also been reported. However, little has been clarified about any related molecular biological mechanisms. We previously revealed that nucleotide substitutions of 377G>A and 2604C>T in the AO gene might be responsible for individual differences in AO activity in Donryu strain rats. By using native polyacrylamide gel electrophoresis/Western blotting in this study, the lack of formation of the AO dimer protein, which is essential for catalytic activity, was shown in poor metabolizer Donryu rats, and this could be a major reason for the individual differences. Rat strain differences were also verified from the same perspectives of nucleotide substitutions and expression levels of a dimer protein. Rat strains with high AO activity showed nucleotide sequences of (377G, 2604C) and a dimer protein. In the case of those with low AO activity, the nucleotide at position 2604 was fixed at T, but varied at position 377, such as G, G/A, and A. An AO dimer was detected in the liver cytosols of rat strains with (377G, 2604T), whereas a monomer was observed in those with (377A, 2604T). These results suggest that the lack of formation of a dimer protein leading to loss of catalytic activity might be due to 377G>A nucleotide substitution. Individual and strain differences in AO activity in rats could be explained by this 377G>A substitution, at least in the rat strains used in this study.
Collapse
Affiliation(s)
- Kunio Itoh
- Department of Drug Metabolism and Pharmacokinetics, Tohoku Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
149
|
Testa B, Krämer SD. The biochemistry of drug metabolism--an introduction: Part 2. Redox reactions and their enzymes. Chem Biodivers 2007; 4:257-405. [PMID: 17372942 DOI: 10.1002/cbdv.200790032] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review continues a general presentation of the metabolism of drugs and other xenobiotics started in a recent issue of Chemistry & Biodiversity. This Part 2 presents the numerous oxidoreductases involved, their nomenclature, relevant biochemical properties, catalytic mechanisms, and the very diverse reactions they catalyze. Many medicinally, environmentally, and toxicologically relevant examples are presented and discussed. Cytochromes P450 occupy a majority of the pages of Part 2, but a large number of relevant oxidoreductases are also considered, e.g., flavin-containing monooxygenases, amine oxidases, molybdenum hydroxylases, peroxidases, and the innumerable dehydrogenases/reductases.
Collapse
Affiliation(s)
- Bernard Testa
- Department of Pharmacy, University Hospital Centre (CHUV), Rue du Bugnon, CH-1011 Lausanne.
| | | |
Collapse
|
150
|
Peretz H, Naamati MS, Levartovsky D, Lagziel A, Shani E, Horn I, Shalev H, Landau D. Identification and characterization of the first mutation (Arg776Cys) in the C-terminal domain of the Human Molybdenum Cofactor Sulfurase (HMCS) associated with type II classical xanthinuria. Mol Genet Metab 2007; 91:23-9. [PMID: 17368066 DOI: 10.1016/j.ymgme.2007.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 02/06/2007] [Accepted: 02/06/2007] [Indexed: 11/28/2022]
Abstract
Classical xanthinuria type II is an autosomal recessive disorder characterized by deficiency of xanthine dehydrogenase and aldehyde oxidase activities due to lack of a common sulfido-olybdenum cofactor (MoCo). Two mutations, both in the N-terminal domain of the Human Molybdenum Cofactor Sulfurase (HMCS), were reported in patients with type II xanthinuria. Whereas the N-terminal domain of HMCS was demonstrated to have cysteine desulfurase activity, the C-terminal domain hypothetically transfers the sulfur to the MoCo. We describe the first mutation in the C-terminal domain of HMCS identified in a Bedouin-Arab child presenting with urolithiasis and in an asymptomatic Jewish female. Patients were diagnosed with type II xanthinuria by homozygosity mapping and/or allopurinol loading test. The Bedouin-Arab child was homozygous for a c.2326C>T (p.Arg776Cys) mutation, while the female patient was compound heterozygous for this and a novel c.1034insA (p.Gln347fsStop379) mutation in the N-terminal domain of HMCS. Cosegregation of the homozygous mutant genotype with hypouricemia and hypouricosuria was demonstrated in the Bedouin family. Haplotype analysis indicated that p.Arg776Cys is a recurrent mutation. Arg776 together with six surrounding amino acid residues were found fully conserved and predicted to be buried in homologous eukaryotic MoCo sulfurases. Moreover, Arg776 is conserved in a diversity of eukaryotic and prokaryotic proteins that posses a domain homologous to the C-terminal domain of HMCS. Our findings suggest that Arg776 is essential for a core structure of the C-terminal domain of the HMCS and identification of a mutation at this site may contribute clarifying the mechanism of MoCo sulfuration.
Collapse
Affiliation(s)
- Hava Peretz
- Clinical Biochemistry Laboratory, Sourasky Medical Center, Tel Aviv, Israel.
| | | | | | | | | | | | | | | |
Collapse
|