1
|
Beers J, Authement AK, Isoherranen N, Jackson KD. Cytosolic Enzymes Generate Cannabinoid Metabolites 7-Carboxycannabidiol and 11-Nor-9-carboxytetrahydrocannabinol. ACS Med Chem Lett 2023; 14:614-620. [PMID: 37197460 PMCID: PMC10184666 DOI: 10.1021/acsmedchemlett.3c00017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
The cannabinoids cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) undergo extensive oxidative metabolism in the liver. Although cytochromes P450 form the primary, pharmacologically active, hydroxylated metabolites of CBD and THC, less is known about the enzymes that generate the major in vivo circulating metabolites of CBD and THC, 7-carboxy-CBD and 11-carboxy-THC, respectively. The purpose of this study was to elucidate the enzymes involved in forming these metabolites. Cofactor dependence experiments with human liver subcellular fractions revealed that 7-carboxy-CBD and 11-carboxy-THC formation is largely dependent on cytosolic NAD+-dependent enzymes, with lesser contributions from NADPH-dependent microsomal enzymes. Experiments with chemical inhibitors provided evidence that 7-carboxy-CBD formation is mainly dependent on aldehyde dehydrogenases and 11-carboxy-THC formation is mediated also in part by aldehyde oxidase. This study is the first to demonstrate the involvement of cytosolic drug-metabolizing enzymes in generating major in vivo metabolites of CBD and THC and addresses a knowledge gap in cannabinoid metabolism.
Collapse
Affiliation(s)
- Jessica
L. Beers
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Aurora K. Authement
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Nina Isoherranen
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Klarissa D. Jackson
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
2
|
Robello M, Barresi E, Baglini E, Salerno S, Taliani S, Settimo FD. The Alpha Keto Amide Moiety as a Privileged Motif in Medicinal Chemistry: Current Insights and Emerging Opportunities. J Med Chem 2021; 64:3508-3545. [PMID: 33764065 PMCID: PMC8154582 DOI: 10.1021/acs.jmedchem.0c01808] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the years, researchers in drug discovery have taken advantage of the use of privileged structures to design innovative hit/lead molecules. The α-ketoamide motif is found in many natural products, and it has been widely exploited by medicinal chemists to develop compounds tailored to a vast range of biological targets, thus presenting clinical potential for a plethora of pathological conditions. The purpose of this perspective is to provide insights into the versatility of this chemical moiety as a privileged structure in drug discovery. After a brief analysis of its physical-chemical features and synthetic procedures to obtain it, α-ketoamide-based classes of compounds are reported according to the application of this motif as either a nonreactive or reactive moiety. The goal is to highlight those aspects that may be useful to understanding the perspectives of employing the α-ketoamide moiety in the rational design of compounds able to interact with a specific target.
Collapse
Affiliation(s)
- Marco Robello
- Synthetic Bioactive Molecules Section, LBC, NIDDK, NIH, 8 Center Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
3
|
Hu Y, Xiao Y, Rao Z, Kumar V, Liu H, Lu C. Carbon-carbon Bond Cleavage Catalyzed by Human Cytochrome P450 Enzymes: α-ketol as the Key Intermediate Metabolite in Sequential Metabolism of Olanexidine. Drug Metab Lett 2019; 14:41-53. [PMID: 31763971 DOI: 10.2174/1872312813666191125095818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Carbon-carbon bond cleavage of a saturated aliphatic moiety is rarely seen in xenobiotic metabolism. Olanexidine (Olanedine®), containing an n-octyl (C8) side chain, was mainly metabolized to various shortened side chain (C4 to C6) acid-containing metabolites in vivo in preclinical species. In liver microsomes and S9, the major metabolites of olanexidine were from multi-oxidation on its n-octyl (C8) side chain. However, the carbon-carbon bond cleavage mechanism of n-octyl (C8) side chain, and enzyme(s) responsible for its metabolism in human remained unknown. METHODS A pair of regioisomers of α-ketol-containing C8 side chain olanexidine analogs (3,2-ketol olanexidine and 2,3-ketol olanexidine) were synthesized, followed by incubation in human liver microsomes, recombinant human cytochrome P450 enzymes or human hepatocytes, and subsequent metabolite identification using LC/UV/MS. RESULTS Multiple shortened side chain (C4 to C6) metabolites were identified, including C4, C5 and C6- acid and C6-hydroxyl metabolites. Among 19 cytochrome P450 enzymes tested, CYP2D6, CYP3A4 and CYP3A5 were identified to catalyze carbon-carbon bond cleavage. CONCLUSION 3,2-ketol olanexidine and 2,3-ketol olanexidine were confirmed as the key intermediates in carbon-carbon bond cleavage. Its mechanism is proposed that a nucleophilic addition of iron-peroxo species, generated by CYP2D6 and CYP3A4/5, to the carbonyl group caused the carbon-carbon bond cleavage between the adjacent hydroxyl and ketone groups. As results, 2,3-ketol olanexidine formed a C6 side chain acid metabolite. While, 3,2-ketol olanexidine formed a C6 side chain aldehyde intermediate, which was either oxidized to a C6 side chain acid metabolite or reduced to a C6 side chain hydroxyl metabolite.
Collapse
Affiliation(s)
- Yiding Hu
- Drug Metabolism and Pharmacokinetics, Sanofi, Waltham, Massachusetts-MA-02451, United States
| | - Yi Xiao
- Department of Pathology and Laboratory Medicine, Children's Hospital, Los Angeles, California-CA 90027, United States
| | - Zhesui Rao
- Drug Metabolism and Pharmacokinetics, Sanofi, Waltham, Massachusetts-MA-02451, United States
| | - Vasant Kumar
- Analytical R&D, Sanofi, Waltham, Massachusetts-MA-02451, United States
| | - Hanlan Liu
- Department of DMPK and Preformulation, KSQ, Cambridge, Massachusetts- MA 02139, United States
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Sanofi, Waltham, Massachusetts-MA-02451, United States
| |
Collapse
|
4
|
Nishimuta H, Watanabe T, Bando K. Quantitative Prediction of Human Hepatic Clearance for P450 and Non-P450 Substrates from In Vivo Monkey Pharmacokinetics Study and In Vitro Metabolic Stability Tests Using Hepatocytes. AAPS JOURNAL 2019; 21:20. [PMID: 30673906 DOI: 10.1208/s12248-019-0294-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/02/2019] [Indexed: 01/01/2023]
Abstract
Accurate prediction of human pharmacokinetics for drugs remains challenging, especially for non-cytochrome P450 (P450) substrates. Hepatocytes might be suitable for predicting hepatic intrinsic clearance (CLint) of new chemical entities, because they can be applied to various compounds regardless of the metabolic enzymes. However, it was reported that hepatic CLint is underestimated in hepatocytes. The purpose of the present study was to confirm the predictability of human hepatic clearance for P450 and non-P450 substrates in hepatocytes and the utility of animal scaling factors for the prediction using hepatocytes. CLint values for 30 substrates of P450, UDP-glucuronosyltransferase, flavin-containing monooxygenase, esterases, reductases, and aldehyde oxidase in human microsomes, human S9 and human, rat, and monkey hepatocytes were estimated. Hepatocytes were incubated in serum of each species. Furthermore, CLint values in human hepatocytes were corrected with empirical, monkey, and rat scaling factors. CLint values in hepatocytes for most compounds were underestimated compared to observed values regardless of the metabolic enzyme, and the predictability was improved by using the scaling factors. The prediction using human hepatocytes corrected with monkey scaling factor showed the highest predictability for both P450 and non-P450 substrates among the predictions using liver microsomes, liver S9, and hepatocytes with or without scaling factors. CLint values by this method for 80% and 90% of all compounds were within 2- and 3-fold of observed values, respectively. This method is accurate and useful for estimating new chemical entities, with no need to care about cofactors, localization of metabolic enzymes, or protein binding in plasma and incubation mixture.
Collapse
Affiliation(s)
- Haruka Nishimuta
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan.
| | - Takao Watanabe
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Kiyoko Bando
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| |
Collapse
|
5
|
Ramsden D, Smith D, Arenas R, Frederick K, Cerny MA. Identification and Characterization of a Selective Human Carbonyl Reductase 1 Substrate. Drug Metab Dispos 2018; 46:1434-1440. [PMID: 30068520 DOI: 10.1124/dmd.118.082487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/26/2018] [Indexed: 02/13/2025] Open
Abstract
During drug discovery efforts targeting inhibition of cytochrome P450 11B2 (CYP11B2)-mediated production of aldosterone as a therapeutic approach for the treatment of chronic kidney disease and hypertension, (S)-6-(5-fluoro-4-(1-hydroxyethyl)pyridin-3-yl)-3,4-dihydro-1,8-naphthyridine-1(2H)-carboxamide (1) was identified as a potent and selective inhibitor of CYP11B2. Preclinical studies characterized 1 as low clearance in both in vitro test systems and in vivo in preclinical species. Despite low metabolic conversion, an active ketone metabolite (2) was identified from in vitro metabolite-identification studies. Due to the inhibitory activity of 2 against CYP11B2 as well as the potential for it to undergo reductive metabolism back to 1, the formation and elimination of 2 were characterized and are the focus of this manuscript. A series of in vitro investigations determined that 1 was slowly oxidized to 2 by cytochrome P450s 2D6, 3A4, and 3A5, followed by stereoselective reduction back to 1 and not its enantiomer (3). Importantly, reduction of 2 was mediated by an NADPH-dependent cytosolic enzyme. Studies with human cytosolic fractions from multiple tissues, selective inhibitors, and recombinantly expressed enzymes indicated that carbonyl reductase 1 (CBR1) is responsible for this transformation in humans. Carbonyl reduction is emerging as an important pathway for endogenous and xenobiotic metabolism. With a lack of selective substrates and inhibitors to enable characterization of the involvement of CBR1, 2 could be a useful probe to assess CBR1 activity in vitro in both subcellular fractions and in cell-based systems.
Collapse
Affiliation(s)
- Diane Ramsden
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Dustin Smith
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Raquel Arenas
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kosea Frederick
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Matthew A Cerny
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
6
|
Shi SM, Di L. The role of carbonyl reductase 1 in drug discovery and development. Expert Opin Drug Metab Toxicol 2017; 13:859-870. [DOI: 10.1080/17425255.2017.1356820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | - Li Di
- Pfizer Inc., Groton, CT, USA
| |
Collapse
|
7
|
Wu X, Zhang Q, Guo J, Jia Y, Zhang Z, Zhao M, Yang Y, Wang B, Hu J, Sheng L, Li Y. Metabolism of F18, a Derivative of Calanolide A, in Human Liver Microsomes and Cytosol. Front Pharmacol 2017; 8:479. [PMID: 28769808 PMCID: PMC5515859 DOI: 10.3389/fphar.2017.00479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/04/2017] [Indexed: 12/02/2022] Open
Abstract
10-Chloromethyl-11-demethyl-12-oxo-calanolide (F18), an analog of calanolide A, is a novel potent nonnucleoside reverse transcriptase inhibitor against HIV-1. Here, we report the metabolic profile and the results of associated biochemical studies of F18 in vitro and in vivo. The metabolites of F18 were identified based on liquid chromatography-electrospray ionization mass spectrometry and/or nuclear magnetic resonance. Twenty-three metabolites of F18 were observed in liver microsomes in vitro. The metabolism of F18 involved 4-propyl chain oxidation, 10-chloromethyl oxidative dechlorination and 12-carbonyl reduction. Three metabolites (M1, M3-1, and M3-2) were also found in rat blood after oral administration of F18 and the reduction metabolites M3-1 and M3-2 were found to exhibit high potency for the inhibition of HIV-1 in vitro. The oxidative metabolism of F18 was mainly catalyzed by cytochrome P450 3A4 in human microsomes, whereas flavin-containing monooxygenases and 11β-hydroxysteroid dehydrogenase were found to be involved in its carbonyl reduction. In human cytosol, multiple carbonyl reductases, including aldo-keto reductase 1C, short-chain dehydrogenases/reductases and quinone oxidoreductase 1, were demonstrated to be responsible for F18 carbonyl reduction. In conclusion, the in vitro metabolism of F18 involves multiple drug metabolizing enzymes, and several metabolites exhibited anti-HIV-1 activities. Notably, the described results provide the first demonstration of the capability of FMOs for carbonyl reduction.
Collapse
Affiliation(s)
- Xiangmeng Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Qinghao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Jiamei Guo
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yufei Jia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Ziqian Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Manman Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yakun Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Li Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| |
Collapse
|
8
|
Maté ML, Geary T, Mackenzie C, Lanusse C, Virkel G. Species differences in hepatic biotransformation of the anthelmintic drug flubendazole. J Vet Pharmacol Ther 2017; 40:493-499. [DOI: 10.1111/jvp.12383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/02/2016] [Indexed: 12/22/2022]
Affiliation(s)
- M. L. Maté
- Laboratorio de Farmacología; Centro de Investigación Veterinaria de Tandil (CIVETAN-CONICET); Facultad de Ciencias Veterinarias; UNCPBA; Tandil Argentina
| | - T. Geary
- Institute of Parasitology; McGill University; Ste-Anne-de-Bellevue QC Canada
| | - C. Mackenzie
- Department of Pathobiology and Diagnostic Investigation; College of Veterinary Medicine; Michigan State University; East Lansing MI USA
| | - C. Lanusse
- Laboratorio de Farmacología; Centro de Investigación Veterinaria de Tandil (CIVETAN-CONICET); Facultad de Ciencias Veterinarias; UNCPBA; Tandil Argentina
| | - G. Virkel
- Laboratorio de Farmacología; Centro de Investigación Veterinaria de Tandil (CIVETAN-CONICET); Facultad de Ciencias Veterinarias; UNCPBA; Tandil Argentina
| |
Collapse
|
9
|
Murata H, Higuchi T, Otagiri M. Oral pharmacokinetics and in-vitro metabolism of metyrapone in male rats. ACTA ACUST UNITED AC 2016; 68:970-9. [PMID: 27265478 DOI: 10.1111/jphp.12560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/29/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate the pharmacokinetics of a single oral administration of metyrapone (MP) and metabolites produced from it in male Wistar rats, and the major tissues and enzymes involved in the production of the MP metabolites. Furthermore, the MP metabolism in human liver subcellular fractions was compared with that in rats. METHODS High-performance liquid chromatography with ultraviolet detection (HPLC-UV) was used to determine the concentrations of MP and its metabolites in plasma and urine after administration, and the production activity of MP metabolites in subcellular fractions of various tissues. KEY FINDINGS Plasma concentration of MP was rapidly increased and decreased, and the primary metabolite, metyrapol (MPOL), was immediately produced. The production activity of MPOL was substantially inhibited by an 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor in the rat and human liver microsomal and mitochondrial fractions. In the liver cytosolic fraction, the activity was inhibited by a carbonyl reductase inhibitor in the humans but not rats. CONCLUSIONS In this study, we elucidated the plasma pharmacokinetics of MP and its metabolites in male rats after an oral administration. MPOL is most likely to be produced by 11β-HSD1 in the male rats and humans.
Collapse
Affiliation(s)
- Hideyuki Murata
- Pre-clinical department, Janssen Pharmaceutical K.K., Chiyoda-ku, Tokyo, Japan.,Nihon Pharmaceutical University, Saitama, Japan
| | | | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Nishi-ku, Kumamoto, Japan
| |
Collapse
|
10
|
Fu ZD, Selwyn FP, Cui JY, Klaassen CD. RNA Sequencing Quantification of Xenobiotic-Processing Genes in Various Sections of the Intestine in Comparison to the Liver of Male Mice. Drug Metab Dispos 2016; 44:842-56. [PMID: 27048750 PMCID: PMC4885488 DOI: 10.1124/dmd.115.068270] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/04/2016] [Indexed: 12/31/2022] Open
Abstract
Previous reports on tissue distribution of xenobiotic-processing genes (XPGs) have limitations, because many non-cytochrome P450 phase I enzymes have not been investigated, and one cannot compare the real mRNA abundance of multiple XPGs using conventional quantification methods. Therefore, this study aimed to quantify and compare the mRNA abundance of all major XPGs in the liver and intestine using RNA sequencing. The mRNA profiles of 304 XPGs, including phase I, phase II enzymes, phase II cosubstrate synthetic enzymes, xenobiotic transporters, as well as xenobiotic-related transcription factors, were systematically examined in the liver and various sections of the intestine in adult male C57BL/6J mice. By two-way hierarchical clustering, over 80% of the XPGs had tissue-divergent expression, which partitioned into liver-predominant, small intestine-predominant, and large intestine-predominant patterns. Among the genes, 54% were expressed highest in the liver, 21% in the duodenum, 4% in the jejunum, 6% in the ileum, and 15% in the large intestine. The highest-expressed XPG in the liver was Mgst1; in the duodenum, Cyp3a11; in the jejunum and ileum, Ces2e; and in the large intestine, Cyp2c55. Interestingly, XPGs in the same family usually exhibited highly different tissue distribution patterns, and many XPGs were almost exclusively expressed in one tissue and minimally expressed in others. In conclusion, the present study is among the first and the most comprehensive investigations of the real mRNA abundance and tissue-divergent expression of all major XPGs in mouse liver and intestine, which aids in understanding the tissue-specific biotransformation and toxicity of drugs and other xenobiotics.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Felcy Pavithra Selwyn
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Curtis D Klaassen
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
11
|
Connarn JN, Zhang X, Babiskin A, Sun D. Metabolism of bupropion by carbonyl reductases in liver and intestine. Drug Metab Dispos 2015; 43:1019-27. [PMID: 25904761 PMCID: PMC6067387 DOI: 10.1124/dmd.115.063107] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022] Open
Abstract
Bupropion's metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. The purpose of this investigation was to compare the relative contribution of the two metabolism pathways of bupropion (by CYP2B6 and CR) in the subcellular fractions of liver and intestine and to identify the CRs responsible for erythro/threohydrobupropion formation in the liver and the intestine. The results showed that the liver microsome generated the highest amount of hydroxybupropion (Vmax = 131 pmol/min per milligram, Km = 87 μM). In addition, liver microsome and S9 fractions formed similar levels of threohydrobupropion by CR (Vmax = 98-99 pmol/min per milligram and Km = 186-265 μM). Interestingly, the liver has similar capability to form hydroxybupropion (by CYP2B6) and threohydrobupropion (by CR). In contrast, none of the intestinal fractions generate hydroxybupropion, suggesting that the intestine does not have CYP2B6 available for metabolism of bupropion. However, intestinal S9 fraction formed threohydrobupropion to the extent of 25% of the amount of threohydrobupropion formed by liver S9 fraction. Enzyme inhibition and Western blots identified that 11β-dehydrogenase isozyme 1 in the liver microsome fraction is mainly responsible for the formation of threohydrobupropion, and in the intestine AKR7 may be responsible for the same metabolite formation. These quantitative comparisons of bupropion metabolism by CR in the liver and intestine may provide new insight into its efficacy and side effects with respect to these metabolites.
Collapse
Affiliation(s)
- Jamie N Connarn
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan (J.N.C., D.S.); Office of Generic Drugs, Food and Drug Administration, Rockville, Maryland (X.Z., A.B.)
| | - Xinyuan Zhang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan (J.N.C., D.S.); Office of Generic Drugs, Food and Drug Administration, Rockville, Maryland (X.Z., A.B.)
| | - Andrew Babiskin
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan (J.N.C., D.S.); Office of Generic Drugs, Food and Drug Administration, Rockville, Maryland (X.Z., A.B.)
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan (J.N.C., D.S.); Office of Generic Drugs, Food and Drug Administration, Rockville, Maryland (X.Z., A.B.)
| |
Collapse
|
12
|
Meng J, Zhong D, Li L, Yuan Z, Yuan H, Xie C, Zhou J, Li C, Gordeev MF, Liu J, Chen X. Metabolism of MRX-I, a novel antibacterial oxazolidinone, in humans: the oxidative ring opening of 2,3-Dihydropyridin-4-one catalyzed by non-P450 enzymes. Drug Metab Dispos 2015; 43:646-59. [PMID: 25710940 DOI: 10.1124/dmd.114.061747] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
MRX-I is an analog of linezolid containing a 2,3-dihydropyridin-4-one (DHPO) ring rather than a morpholine ring. Our objectives were to characterize the major metabolic pathways of MRX-I in humans and clarify the mechanism underlying the oxidative ring opening of DHPO. After an oral dose of MRX-I (600 mg), nine metabolites were identified in humans. The principal metabolic pathway proposed involved the DHPO ring opening, generating the main metabolites in the plasma and urine: the hydroxyethyl amino propionic acid metabolite MRX445-1 and the carboxymethyl amino propionic acid metabolite MRX459. An in vitro phenotyping study demonstrated that multiple non-cytochrome P450 enzymes are involved in the formation of MRX445-1 and MRX459, including flavin-containing monooxygenase 5, short-chain dehydrogenase/reductase, aldehyde ketone reductase, and aldehyde dehydrogenase (ALDH). H2 (18)O experiments revealed that two (18)O atoms are incorporated into MRX445-1, one in the carboxyethyl group and the other in the hydroxyl group, and three (18)O atoms are incorporated into MRX459, two in the carboxymethyl group and one in the hydroxyl group. Based on these results, the mechanism proposed for the DHPO ring opening involves the metabolism of MRX-I via FMO5-mediated Baeyer-Villiger oxidation to an enol lactone, hydrolysis to an enol, and enol-aldehyde tautomerism to an aldehyde. The aldehyde is reduced by short-chain dehydrogenase/reductase, aldehyde ketone reductase, ALDH to MRX445-1, or oxidized by ALDH to MRX459. Our study suggests that few clinical adverse drug-drug interactions should be anticipated between MRX-I and cytochrome P450 inhibitors or inducers.
Collapse
Affiliation(s)
- Jian Meng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Dafang Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Liang Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Zhengyu Yuan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Hong Yuan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Cen Xie
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Jialan Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Chen Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Mikhail Fedorovich Gordeev
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Jinqian Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.M., D.Z., L.L., C.X., J.Z., C.L., X.C.); MicuRx Pharmaceuticals, Inc., Shanghai, China (Z.Y., H.Y.); and MicuRx Pharmaceuticals, Inc., Hayward, California (M.G., J.L.)
| |
Collapse
|
13
|
Andrýs R, Zemanová L, Lenčo J, Bílková Z, Wsól V. Carbonyl-reducing enzymes as targets of a drug-immobilised affinity carrier. Chem Biol Interact 2015; 234:169-77. [PMID: 25595226 DOI: 10.1016/j.cbi.2015.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 12/20/2014] [Accepted: 01/02/2015] [Indexed: 01/21/2023]
Abstract
Proteins, peptides and nucleic acids are commonly isolated and purified in almost all bioscience laboratories. Methods based on molecular recognition are currently the most powerful tool in separation processes due to their selectivity and recovery. The aim of this study was to prove the versatility and the ability of an affinity carrier containing the immobilised ligand oracin (previously developed by our workgroup) to selectively bind carbonyl-reducing enzymes. These enzymes play an important role in metabolic pathways of various endogenic compounds and xenobiotics. Many important drugs, such as doxorubicin, daunorubicin, haloperidol and the model anticancer drug oracin, are metabolised by carbonyl-reducing enzymes. The functionality of the presented carrier was demonstrated with pure recombinant enzymes (AKR1A1, AKR1B1, AKR1B10, AKR1C1, AKR1C2, AKR1C3, AKR1C4, CBR1 and CBR3) as well as with two model biological samples (cell extract from genetically modified Escherichia coli and pre-purified human liver cytosol). Enzymes that show an affinity toward oracin were efficiently captured, gently eluted using 150 mM ammonium hydroxide and subsequently identified by MS. The method is highly selective and robust and may be applied to the purification and identification of various carbonyl-reducing enzymes from any biological sample.
Collapse
Affiliation(s)
- Rudolf Andrýs
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, Hradec Králové 500 05, Czech Republic.
| | - Lucie Zemanová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, Hradec Králové 500 05, Czech Republic.
| | - Juraj Lenčo
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, Hradec Králové 500 05, Czech Republic; Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Třebešská 1575, Hradec Králové 500 01, Czech Republic.
| | - Zuzana Bílková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, Pardubice 532 10, Czech Republic.
| | - Vladimír Wsól
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, Hradec Králové 500 05, Czech Republic.
| |
Collapse
|
14
|
Alshogran OY, Naud J, Ocque AJ, Leblond FA, Pichette V, Nolin TD. Effect of experimental kidney disease on the functional expression of hepatic reductases. Drug Metab Dispos 2015; 43:100-6. [PMID: 25332430 DOI: 10.1124/dmd.114.061150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Chronic kidney disease (CKD) affects the nonrenal clearance of drugs by modulating the functional expression of hepatic drug-metabolizing enzymes and transporters. The impact of CKD on oxidative and conjugative metabolism has been extensively studied. However, its effect on hepatic drug reduction, an important phase I drug-metabolism pathway, has not been investigated. We aimed to assess the effect of experimental CKD on hepatic reduction using warfarin as a pharmacological probe substrate. Cytosolic and microsomal cellular fractions were isolated from liver tissue harvested from five-sixths-nephrectomized and control rats (n = 10 per group). The enzyme kinetics for warfarin reduction were evaluated in both fractions, and formation of warfarin alcohols was used as an indicator of hepatic reductase activity. Selective inhibitors were employed to identify reductases involved in warfarin reduction. Gene and protein expression of reductases were determined using quantitative real-time polymerase chain reaction and Western blotting, respectively. Formation of RS/SR-warfarin alcohol was decreased by 39% (P < 0.001) and 43% (P < 0.01) in cytosol and microsomes, respectively, in CKD rats versus controls. However, RR/SS-warfarin alcohol formation was unchanged in the cytosol, and a trend toward its decreased production was observed in microsomes. Gene and protein expression of cytosolic carbonyl reductase 1 and aldo-keto reductase 1C3/18, and microsomal 11β-hydroxysteroid dehydrogenase type 1 were significantly reduced by >30% (P < 0.05) in CKD rats compared with controls. Collectively, these results suggest that the functional expression of hepatic reductases is selectively decreased in kidney disease. Our findings may explain one mechanism for altered nonrenal clearance, exposure, and response of drugs in CKD patients.
Collapse
Affiliation(s)
- Osama Y Alshogran
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| | - Judith Naud
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| | - Andrew J Ocque
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| | - François A Leblond
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| | - Vincent Pichette
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| | - Thomas D Nolin
- Center for Clinical Pharmaceutical Sciences (O.Y.A., A.J.O., T.D.N.), Department of Pharmaceutical Sciences (O.Y.A.) and Department of Pharmacy and Therapeutics (T.D.N.), School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania; and Service de Néphrologie et Centre de Recherche, Hôpital Maisonneuve-Rosemont (J.N., F.A.L., V.P.), Département de Pharmacologie (V.P.), Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
15
|
Alleman RJ, Katunga LA, Nelson MAM, Brown DA, Anderson EJ. The "Goldilocks Zone" from a redox perspective-Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front Physiol 2014; 5:358. [PMID: 25278906 PMCID: PMC4166897 DOI: 10.3389/fphys.2014.00358] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 01/17/2023] Open
Abstract
Consequences of oxidative stress may be beneficial or detrimental in physiological systems. An organ system's position on the “hormetic curve” is governed by the source and temporality of reactive oxygen species (ROS) production, proximity of ROS to moieties most susceptible to damage, and the capacity of the endogenous cellular ROS scavenging mechanisms. Most importantly, the resilience of the tissue (the capacity to recover from damage) is a decisive factor, and this is reflected in the disparate response to ROS in cardiac and skeletal muscle. In myocytes, a high oxidative capacity invariably results in a significant ROS burden which in homeostasis, is rapidly neutralized by the robust antioxidant network. The up-regulation of key pathways in the antioxidant network is a central component of the hormetic response to ROS. Despite such adaptations, persistent oxidative stress over an extended time-frame (e.g., months to years) inevitably leads to cumulative damages, maladaptation and ultimately the pathogenesis of chronic diseases. Indeed, persistent oxidative stress in heart and skeletal muscle has been repeatedly demonstrated to have causal roles in the etiology of heart disease and insulin resistance, respectively. Deciphering the mechanisms that underlie the divergence between adaptive and maladaptive responses to oxidative stress remains an active area of research for basic scientists and clinicians alike, as this would undoubtedly lead to novel therapeutic approaches. Here, we provide an overview of major types of ROS in striated muscle and the divergent adaptations that occur in response to them. Emphasis is placed on highlighting newly uncovered areas of research on this topic, with particular focus on the mitochondria, and the diverging roles that ROS play in muscle health (e.g., exercise or preconditioning) and disease (e.g., cardiomyopathy, ischemia, metabolic syndrome).
Collapse
Affiliation(s)
- Rick J Alleman
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Lalage A Katunga
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Margaret A M Nelson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Ethan J Anderson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
16
|
Wu Y, Liu ZY, Cao Y, Chen XJ, Zeng JG, Sun ZL. Reductive metabolism of the sanguinarine iminium bond by rat liver preparations. Pharmacol Rep 2014; 65:1391-400. [PMID: 24399736 DOI: 10.1016/s1734-1140(13)71498-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 08/13/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sanguinarine (SA) is a quaternary benzo[c]phenanthridine alkaloid that is mainly present in the Papaveraceae family. SA has been extensively studied because of its antimicrobial, anti-inflammatory, antitumor, antihypertensive, antiproliferative and antiplatelet activities. Metabolic studies demonstrated that SA bioavailability is apparently low, and the main pathway of SA metabolism is iminium bond reduction resulting in dihydrosanguinarine (DHSA) formation. Nevertheless, the metabolic enzymes involved in SA reduction are still not known in detail. Thus, the aim of this study was to investigate the rat liver microsomes and cytosol-induced SA iminium bond reduction, and to examine the effects of cytosol reductase inhibitors on the reductive activity. METHODS DHSA formation was quantified by HPLC. The possible enzymes responsible for DHSA formation were examined using selective individual metabolic enzyme inhibitors. RESULTS When SA was incubated with liver microsomes and cytosol in the absence of NAD(P)H, DHSA, the iminium bond reductive metabolite was formed. The reductase activity of the liver microsomes and cytosol was also enhanced significantly in the presence of NADH. The amount of DHSA formed in the liver cytosol was 4.6-fold higher than in the liver microsomes in the presence of NADH. The reductase activity in the liver cytosol was inhibited by the addition of flavin mononucleotide and/or riboflavin. Inhibition studies indicated that menadione, dicoumarol, quercetin and 7-hydroxycoumarin inhibited rat liver cytosol-mediated DHSA formation in the absence of NADH. However, only menadione and quercetin inhibited rat liver cytosol-mediated DHSA formation in the presence of NADH. CONCLUSIONS These results suggest that the SA iminium bond reduction proceeds via two routes in the liver cytosol. One route is direct non-enzymatic reduction by NAD(P)H, and the other is enzymatic reduction by possible carbonyl and/or quinone reductases in the liver cytosol.
Collapse
Affiliation(s)
- Yong Wu
- Hunan Engineering Research Center of Veterinary Drug, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | | | | | | | | | | |
Collapse
|
17
|
Larsen K, Najle R, Lifschitz A, Maté ML, Lanusse C, Virkel GL. Effects of Sublethal Exposure to a Glyphosate-Based Herbicide Formulation on Metabolic Activities of Different Xenobiotic-Metabolizing Enzymes in Rats. Int J Toxicol 2014; 33:307-318. [PMID: 24985121 DOI: 10.1177/1091581814540481] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The activities of different xenobiotic-metabolizing enzymes in liver subcellular fractions from Wistar rats exposed to a glyphosate (GLP)-based herbicide (Roundup full II) were evaluated in this work. Exposure to the herbicide triggered protective mechanisms against oxidative stress (increased glutathione peroxidase activity and total glutathione levels). Liver microsomes from both male and female rats exposed to the herbicide had lower (45%-54%, P < 0.01) hepatic cytochrome P450 (CYP) levels compared to their respective control animals. In female rats, the hepatic 7-ethoxycoumarin O-deethylase (a general CYP-dependent enzyme activity) was 57% higher (P < 0.05) in herbicide-exposed compared to control animals. Conversely, this enzyme activity was 58% lower (P < 0.05) in male rats receiving the herbicide. Lower (P < 0.05) 7-ethoxyresorufin O-deethlyase (EROD, CYP1A1/2 dependent) and oleandomycin triacetate (TAO) N-demethylase (CYP3A dependent) enzyme activities were observed in liver microsomes from exposed male rats. Conversely, in females receiving the herbicide, EROD increased (123%-168%, P < 0.05), whereas TAO N-demethylase did not change. A higher (158%-179%, P < 0.01) benzyloxyresorufin O-debenzylase (a CYP2B-dependent enzyme activity) activity was only observed in herbicide-exposed female rats. In herbicide-exposed rats, the hepatic S-oxidation of methimazole (flavin monooxygenase dependent) was 49% to 62% lower (P < 0.001), whereas the carbonyl reduction of menadione (a cytosolic carbonyl reductase-dependent activity) was higher (P < 0.05). Exposure to the herbicide had no effects on enzymatic activities dependent on carboxylesterases, glutathione transferases, and uridinediphospho-glucuronosyltransferases. This research demonstrated certain biochemical modifications after exposure to a GLP-based herbicide. Such modifications may affect the metabolic fate of different endobiotic and xenobiotic substances. The pharmacotoxicological significance of these findings remains to be clarified.
Collapse
Affiliation(s)
- Karen Larsen
- Laboratorio de Biología y Ecotoxicología, Facultad de Ciencias Veterinarias, UNCPBA, Tandil, Argentina
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias (UNCPBA), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET), Tandil, Argentina
| | - Roberto Najle
- Laboratorio de Biología y Ecotoxicología, Facultad de Ciencias Veterinarias, UNCPBA, Tandil, Argentina
| | - Adrián Lifschitz
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias (UNCPBA), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET), Tandil, Argentina
| | - María L Maté
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias (UNCPBA), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET), Tandil, Argentina
| | - Carlos Lanusse
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias (UNCPBA), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET), Tandil, Argentina
| | - Guillermo L Virkel
- Laboratorio de Farmacología, Facultad de Ciencias Veterinarias (UNCPBA), Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET), Tandil, Argentina
| |
Collapse
|
18
|
Interindividual variability in the cardiac expression of anthracycline reductases in donors with and without Down syndrome. Pharm Res 2014; 31:1644-55. [PMID: 24562808 DOI: 10.1007/s11095-013-1267-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
PURPOSE The intracardiac synthesis of anthracycline alcohol metabolites (e.g., daunorubicinol) contributes to the pathogenesis of anthracycline-related cardiotoxicity. Cancer patients with Down syndrome (DS) are at increased risk for anthracycline-related cardiotoxicity. We profiled the expression of anthracycline metabolizing enzymes in hearts from donors with- and without- DS. METHODS Cardiac expression of CBR1, CBR3, AKR1A1, AKR1C3 and AKR7A2 was examined by quantitative real time PCR, quantitative immunoblotting, and enzyme activity assays using daunorubicin. The CBR1 polymorphism rs9024 was investigated by allelic discrimination with fluorescent probes. The contribution of CBRs/AKRs proteins to daunorubicin reductase activity was examined by multiple linear regression. RESULTS CBR1 was the most abundant transcript (average relative expression; DS: 81%, non-DS: 58%), and AKR7A2 was the most abundant protein (average relative expression; DS: 38%, non-DS: 35%). Positive associations between cardiac CBR1 protein levels and daunorubicin reductase activity were found for samples from donors with- and without- DS. Regression analysis suggests that sex, CBR1, AKR1A1, and AKR7A2 protein levels were significant contributors to cardiac daunorubicin reductase activity. CBR1 rs9024 genotype status impacts on cardiac CBR1 expression in non-DS hearts. CONCLUSIONS CBR1, AKR1A1, and AKR7A2 protein levels point to be important determinants for predicting the synthesis of cardiotoxic daunorubicinol in heart.
Collapse
|
19
|
|
20
|
|
21
|
Nishimuta H, Nakagawa T, Nomura N, Yabuki M. Significance of reductive metabolism in human intestine and quantitative prediction of intestinal first-pass metabolism by cytosolic reductive enzymes. Drug Metab Dispos 2013; 41:1104-11. [PMID: 23444387 DOI: 10.1124/dmd.113.051177] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The number of new drug candidates that are cleared via non-cytochrome P450 (P450) enzymes has increased. However, unlike oxidation by P450, the roles of reductive enzymes are less understood. The metabolism in intestine is especially not well known. The purposes of this study were to investigate the significance of reductive metabolism in human intestine, and to establish a quantitative prediction method of intestinal first-pass metabolism by cytosolic reductive enzymes, using haloperidol, mebendazole, and ziprasidone. First, we estimated the metabolic activities for these compounds in intestine and liver using subcellular fractions. Metabolic activities were detected in human intestinal cytosol (HIC) for all three compounds, and the intrinsic clearance values were higher than those in human liver cytosol for haloperidol and mebendazole. These metabolic activities in HIC were NADPH- and/or NADH-dependent. Furthermore, the metabolic activities for all three compounds in HIC were largely inhibited by menadione, which has been used as a carbonyl reductase (CBR)-selective chemical inhibitor. Therefore, considering subcellular location, cofactor requirement, and chemical inhibition, these compounds might be metabolized by CBRs in human intestine. Subsequently, we tried to quantitatively predict intestinal availability (F(g)) for these compounds using human intestinal S9 (HIS9). Our prediction model using apparent permeability of parallel artificial membrane permeability assay and metabolic activities in HIS9 could predict F(g) in humans for the three compounds well. In conclusion, CBRs might have higher metabolic activities in human intestine than in human liver. Furthermore, our prediction method of human F(g) using HIS9 is applicable to substrates of cytosolic reductive enzymes.
Collapse
Affiliation(s)
- Haruka Nishimuta
- Pharmacokinetics Research Laboratory, Dainippon Sumitomo Pharma Co., Ltd., Enoki 33-94, Suita, Osaka 564-0053, Japan.
| | | | | | | |
Collapse
|
22
|
Škarydová L, Andrýs R, Holubová L, Štambergová H, Kňavová J, Wsól V, Bílková Z. Efficient isolation of carbonyl-reducing enzymes using affinity approach with anticancer drug oracin as a specific ligand. J Sep Sci 2013; 36:1176-84. [DOI: 10.1002/jssc.201201141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/13/2013] [Accepted: 01/13/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Lucie Škarydová
- Department of Biochemical Sciences; Faculty of Pharmacy in Hradec Králové; Charles University in Prague; Hradec Králové Czech Republic
| | - Rudolf Andrýs
- Department of Biochemical Sciences; Faculty of Pharmacy in Hradec Králové; Charles University in Prague; Hradec Králové Czech Republic
| | - Lucie Holubová
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology; University of Pardubice; Pardubice Czech Republic
| | - Hana Štambergová
- Department of Biochemical Sciences; Faculty of Pharmacy in Hradec Králové; Charles University in Prague; Hradec Králové Czech Republic
| | - Jana Kňavová
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology; University of Pardubice; Pardubice Czech Republic
| | - Vladimír Wsól
- Department of Biochemical Sciences; Faculty of Pharmacy in Hradec Králové; Charles University in Prague; Hradec Králové Czech Republic
| | - Zuzana Bílková
- Department of Biological and Biochemical Sciences; Faculty of Chemical Technology; University of Pardubice; Pardubice Czech Republic
| |
Collapse
|
23
|
Skarydova L, Nobilis M, Wsól V. Role of carbonyl reducing enzymes in the phase I biotransformation of the non-steroidal anti-inflammatory drug nabumetone in vitro. Xenobiotica 2012; 43:346-54. [PMID: 23020786 DOI: 10.3109/00498254.2012.720048] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. Nabumetone is a clinically used non-steroidal anti-inflammatory drug, its biotransformation includes major active metabolite 6-methoxy-2-naphtylacetic acid and another three phase I as well as corresponding phase II metabolites which are regarded as inactive. One important biotransformation pathway is carbonyl reduction, which leads to the phase I metabolite, reduced nabumetone. 2. The aim of this study is the determination of the role of a particular human liver subcellular fraction in the nabumetone reduction and the identification of participating carbonyl reducing enzymes along with their stereospecificities. 3. Both subcellular fractions take part in the carbonyl reduction of nabumetone and the reduction is at least in vitro the main biotransformation pathway. The activities of eight cytosolic carbonyl reducing enzymes--CBR1, CBR3, AKR1B1, AKR1B10, AKR1C1-4--toward nabumetone were tested. Except for CBR3, all tested reductases transform nabumetone to its reduced metabolite. AKR1C4 and AKR1C3 have the highest intrinsic clearances. 4. The stereospecificity of the majority of the tested enzymes is shifted to the production of an (+)-enantiomer of reduced nabumetone; only AKR1C1 and AKR1C4 produce predominantly an (-)-enantiomer. This project provides for the first time evidence that seven specific carbonyl reducing enzymes participate in nabumetone metabolism.
Collapse
Affiliation(s)
- Lucie Skarydova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Czech Republic
| | | | | |
Collapse
|
24
|
Abstract
Bupropion is metabolized extensively in humans by oxidative and reductive processes. CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. The objective of this study was to examine the enzyme kinetics of bupropion reduction in human liver. In human liver cytosol, the reduction of bupropion to erythro-and threohydrobupropion was NADPH dependent with Cl(int) values of 0.08 and 0.60 µL·min(-1)mg(-1) protein, respectively. Bupropion reduction in liver microsomes was also NADPH dependent with Cl(int) values of 10.4 and 280 µL·min(-1)mg(-1) protein, respectively. Formation of erythro-and threohydrobupropion in microsomes exceeded that in cytosol by 70 and 170 fold, respectively. Menadione, an inhibitor of cytosolic carbonyl reducing enzymes (e.g. CBRs), inhibited erythro-and threohydrobupropion formation in cytosol with IC(50) of 30 and 54 µM, respectively. In microsomes 18β-glycyrrhetinic acid, an inhibitor of microsomal carbonyl reductases (e.g. 11β-HSDs), inhibited their formation with IC(50) of 25 and 26 nM, respectively. Our findings, in agreement with recent human placental studies, show that carbonyl reducing enzymes in hepatic microsomes are significant players in bupropion reduction. Contrary to past studies, we found that threohydrobupropion (not hydroxybupropion) is the major microsomal generated hepatic metabolite of bupropion.
Collapse
Affiliation(s)
- Jillissa C Molnari
- Department of Pharmaceutical, Biomedical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, Des Moines, IA, USA
| | | |
Collapse
|
25
|
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: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Škarydová L, Wsól V. Human microsomal carbonyl reducing enzymes in the metabolism of xenobiotics: well-known and promising members of the SDR superfamily. Drug Metab Rev 2011; 44:173-91. [DOI: 10.3109/03602532.2011.638304] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
27
|
Skarka A, Škarydová L, Štambergová H, Wsól V. Anthracyclines and their metabolism in human liver microsomes and the participation of the new microsomal carbonyl reductase. Chem Biol Interact 2011; 191:66-74. [DOI: 10.1016/j.cbi.2010.12.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 12/15/2010] [Accepted: 12/16/2010] [Indexed: 01/24/2023]
|
28
|
Hijaz F, Shrestha TB, Bossman SH, Hussain F, Smith JS. In vitro and in vivo metabolism of the radiolytic compound 2-dodecylcyclobutanone. J Food Sci 2010; 75:T72-80. [PMID: 20546430 DOI: 10.1111/j.1750-3841.2010.01600.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Our knowledge about the metabolism of alkylcyclobutanones (2-ACBs) is limited, and the lack of literature on the metabolism of 2-ACBs causes consumers to doubt the safety of irradiated foods. The objectives of this study were to evaluate the metabolism of 2-dodecylcyclobutanone (2-DCB) and identify any possible metabolite. The 2-DCB was mixed with rat S9 (postmitochondrial supernatant fraction) and beta-nicotinamide adenine dinucleotide phosphate (NADPH) in phosphate buffer (pH 7.4) and incubated for 2 h at 37 degrees C. Then, the incubation mixture was mixed with sodium sulfate and extracted with n-hexane by using a Soxhlet apparatus. The hexane extract was concentrated under nitrogen and injected into the gas chromatography-mass spectrometry (GC-MS) machine running in selective ion monitoring mode (SIM) to measure 2-DCB concentration. The hexane extract from the in vitro and in vivo studies was also derivatized with a silylation reagent and injected into a GC-MS running in full scan mode. The average percentage of 2-DCB recovered from the test incubations was 23%, compared with 50% from the controls. The GC-MS chromatograms of the derivatized samples showed a unique peak in the in vitro test incubations and in the hexane extract of the rat feces that were given 2-DCB. This peak was later identified as 2-doecylcyclobutanol.
Collapse
Affiliation(s)
- Faraj Hijaz
- Food Science Inst., Dept. of Animal Sciences and Industry, 208 Call Hall, Kansas State Univ., Manhattan, KS 66506, USA
| | | | | | | | | |
Collapse
|
29
|
Huang W, Ding L, Huang Q, Hu H, Liu S, Yang X, Hu X, Dang Y, Shen S, Li J, Ji X, Jiang S, Liu JO, Yu L. Carbonyl reductase 1 as a novel target of (-)-epigallocatechin gallate against hepatocellular carcinoma. Hepatology 2010; 52:703-14. [PMID: 20683966 DOI: 10.1002/hep.23723] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
UNLABELLED Human carbonyl reductase 1 (CBR1) converts the antitumor drug and anthracycline daunorubicin (DNR) into the alcohol metabolite daunorubicinol (DNROL) with significantly reduced antitumor activity and cardiotoxicity, and this limits the clinical use of DNR. Inhibition of CBR1 can thus increase the efficacy and decrease the toxicity of DNR. Here we report that (-)-epigallocatechin gallate (EGCG) from green tea is a promising inhibitor of CBR1. EGCG directly interacts with CBR1 and acts as a noncompetitive inhibitor with respect to the cofactor reduced nicotinamide adenine dinucleotide phosphate and the substrate isatin. The inhibition is dependent on the pH, and the gallate moiety of EGCG is required for activity. Molecular modeling has revealed that EGCG occupies the active site of CBR1. Furthermore, EGCG specifically enhanced the antitumor activity of DNR against hepatocellular carcinoma SMMC7721 cells expressing high levels of CBR1 and corresponding xenografts. We also demonstrated that EGCG could overcome the resistance to DNR by Hep3B cells stably expressing CBR1 but not by RNA interference of CBR1-HepG2 cells. The level of the metabolite DNROL was negatively correlated with that of EGCG in the cell extracts. Finally, EGCG decreased the cardiotoxicity of DNR in a human carcinoma xenograft model with both SMMC7721 and Hep3B cells in mice. CONCLUSION These results strongly suggest that EGCG can inhibit CBR1 activity and enhance the effectiveness and decrease the cardiotoxicity of the anticancer drug DNR. These findings also indicate that a combination of EGCG and DNR might represent a novel approach for hepatocellular carcinoma therapy or chemoprevention.
Collapse
Affiliation(s)
- Weixue Huang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wang X, Abdelrahman DR, Zharikova OL, Patrikeeva SL, Hankins GDV, Ahmed MS, Nanovskaya TN. Bupropion metabolism by human placenta. Biochem Pharmacol 2010; 79:1684-90. [PMID: 20109440 PMCID: PMC2847018 DOI: 10.1016/j.bcp.2010.01.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 11/23/2022]
Abstract
Smoking during pregnancy is the largest modifiable risk factor for pregnancy-related morbidity and mortality. The success of bupropion for smoking cessation warrants its investigation for the treatment of pregnant patients. Nevertheless, the use of bupropion for the treatment of pregnant smokers requires additional data on its bio-disposition during pregnancy. Therefore, the aim of this investigation was to determine the metabolism of bupropion in placentas obtained from nonsmoking and smoking women, identify metabolites formed and the enzymes catalyzing their formation, as well as the kinetics of the reaction. Data obtained revealed that human placentas metabolized bupropion to hydroxybupropion, erythro- and threohydrobupropion. The rates for formation of erythro- and threohydrobupropion exceeded that for hydroxybupropion by several folds, were dependent on the concentration of bupropion and exhibited saturation kinetics with an apparent K(m) value of 40microM. Human placental 11beta-hydroxysteroid dehydrogenases were identified as the major carbonyl-reducing enzymes responsible for the reduction of bupropion to threo- and erythrohydrobupropion in microsomal fractions. On the other hand, CYP2B6 was responsible for the formation of OH-bupropion. These data suggest that both placental microsomal carbonyl-reducing and oxidizing enzymes are involved in the metabolism of bupropion.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 77555-0587, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Tong Z, Chandrasekaran A, Li H, Rotshteyn Y, Erve JCL, Demaio W, Talaat R, Hultin T, Scatina J. In vitro metabolism and identification of human enzymes involved in the metabolism of methylnaltrexone. Drug Metab Dispos 2010; 38:801-7. [PMID: 20173089 DOI: 10.1124/dmd.110.032169] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Methylnaltrexone (MNTX) is a peripherally acting mu-opioid receptor antagonist and is currently indicated for the treatment of opioid-induced constipation in patients with advanced illness who are receiving palliative care, when response to laxative therapy has not been sufficient. Sulfation to MNTX-3-sulfate (M2) and carbonyl reduction to methyl-6alpha-naltrexol (M4) and methyl-6beta-naltrexol (M5) are the primary metabolic pathways for MNTX in humans. The objectives of this study were to investigate MNTX in vitro metabolism in human and nonclinical species and to identify the human enzymes involved in MNTX metabolism. Of the five commercially available sulfotransferases investigated, only SULT2A1 and SULT1E1 catalyzed M2 formation. Formation of M4 and M5 was catalyzed by NADPH-dependent hepatic cytosolic enzymes, which were identified using selective chemical inhibitors (10 and 100 microM) for aldo-keto reductase (AKR) isoforms, short-chain dehydrogenase/reductase including carbonyl reductase, alcohol dehydrogenase, and quinone oxidoreductase. The results were then compared with the effects of the same inhibitors on 6beta-naltrexol formation from naltrexone, a structural analog of MNTX, which is catalyzed mainly by AKR1C4. The AKR1C inhibitor phenolphthalein inhibited MNTX and naltrexone reduction up to 98%. 5beta-Cholanic acid 3alpha,7alpha-diol, the AKR1C2 inhibitor, and medroxyprogesterone acetate, an inhibitor of AKR1C1, AKR1C2, and AKR1C4, inhibited MNTX reduction up to 67%. Other inhibitors were less potent. In conclusion, the carbonyl reduction of MNTX to M4 and M5 in hepatic cytosol was consistent with previous in vivo observations. AKR1C4 appeared to play a major role in the carbonyl reduction of MNTX, although multiple enzymes in the AKR1C subfamily may be involved. Human SULT2A1 and SULT1E1 were involved in MNTX sulfation.
Collapse
Affiliation(s)
- Zeen Tong
- Pfizer Inc., Collegeville, PA 19426, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
MATÉ L, VIRKEL G, LIFSCHITZ A, SALLOVITZ J, BALLENT M, LANUSSE C. Phase 1 and phase 2 metabolic activities along the small intestine in adult male sheep1. J Vet Pharmacol Ther 2010; 33:537-45. [DOI: 10.1111/j.1365-2885.2010.01177.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Blech S, Ludwig-Schwellinger E, Gräfe-Mody EU, Withopf B, Wagner K. The metabolism and disposition of the oral dipeptidyl peptidase-4 inhibitor, linagliptin, in humans. Drug Metab Dispos 2010; 38:667-78. [PMID: 20086031 DOI: 10.1124/dmd.109.031476] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pharmacokinetics and metabolism of linagliptin (BI1356, 8-(3R-amino-piperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione) were investigated in healthy volunteers. The 10- and 5-mg (14)C-labeled drug was administered orally or intravenously, respectively. Fecal excretion was the dominant excretion pathway with 84.7% (p.o.) and 58.2% (i.v.) of the dose. Renal excretion accounted for 5.4% (p.o.) and 30.8% (i.v.) of the dose. Unchanged linagliptin was the most abundant radioactive species in all matrices investigated. The exposure (area under the curve 0-24 h) to the parent compound in plasma accounted for 191 nM . h (p.o.) and 356 nM . h (i.v.), respectively. The main metabolite 7-but-2-ynyl-8-(3S-hydroxy-piperidin-1-yl)-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione (CD1790) was observed with >10% of parent compound systemic exposure after oral administration. The metabolite was identified as S-3-hydroxypiperidinly derivative of linagliptin. Experiments that included stable-labeled isotope techniques indicated that CD1790 was formed by a two-step mechanism via the ketone 7-but-2-yn-1-yl-3-methyl-1-[(4-methylquinazolin-2-yl)methyl]-8-(3-oxopiperidin-1-yl)-3,7-dihydro-1H-purine-2,6-dione (CD10604). The initial ketone formation was CYP3A4-dependent and rate-limiting for the overall reaction to CD1790. Aldo-keto reductases with minor contribution of carbonyl reductases were involved in the subsequent stereoselective reduction of CD10604 to CD1790. The antipodes of linagliptin and CD1790 were not observed with adequate enantioselective liquid chromatography-tandem mass spectrometry methods. Other minor metabolites were identified by mass spectrometry and NMR investigations. However, it was concluded that the metabolites of linagliptin only play a minor role in the overall disposition and elimination of linagliptin.
Collapse
Affiliation(s)
- Stefan Blech
- Department of Drug Metabolism, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| | | | | | | | | |
Collapse
|
34
|
Li C, Lu B, Garbaccio RM, Tasber ES, Fraley ME, Hartman GD, Ye J, Harrelson JC, Prueksaritanont T. Stereospecific reduction of a potent kinesin spindle protein (KSP) inhibitor in human tissues. Biochem Pharmacol 2010; 79:1526-33. [PMID: 20109439 DOI: 10.1016/j.bcp.2010.01.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 01/19/2010] [Accepted: 01/20/2010] [Indexed: 11/27/2022]
Abstract
Compound A, 1-{(3R,3aR)-3-[3-(4-acetylpiperazin-1-yl)propyl]-7-fluoro-3-phenyl-3a,4-dihydro-3H-pyrazolo[5,1-c][1,4]benzoxazin-2-yl}ethanone, is a novel and potent inhibitor of the mitotic kinesin spindle protein. Metabolism studies with human hepatocytes showed that Compound A underwent significant ketone reduction to its biologically active metabolite M1. Here, we describe the studies that characterized the metabolic interconversion between Compound A and M1 in vitro in human tissues. LC-MS/MS analysis showed that the ketone reduction was stereospecific for M1 with no diastereomer of M1 detected in incubations with human hepatocytes. Interestingly, such stereospecific ketone reduction was not observed with Compound B, the enantiomer of Compound A. No reductive products were observed when Compound B was incubated with human hepatocytes. Studies with human liver subcellular fractions showed that Compound A was reduced to M1 primarily by human liver cytosol with little contribution from human liver microsomes and mitochondria. NADPH was the preferred cofactor for the reduction reaction. Reverse oxidation of M1 back to Compound A was also observed, preferentially in human liver cytosol with NADP(+) as the cofactor. The interconversion between Compound A and M1 in human liver cytosol was inhibited significantly by flufenamic acid and phenolphthalein (potent inhibitors for aldo-keto reductase 1Cs, p<0.05), but not by menadione, a selective inhibitor for carbonyl reductase. In addition to the liver, S9 from human lung and kidney was also capable of catalyzing this interconversion. Collectively, the results implicated the aldo-keto reductase 1Cs as the most likely enzymes responsible for the metabolic interconversion of Compound A and its active metabolite M1.
Collapse
Affiliation(s)
- Chunze Li
- Department of Drug Metabolism and Pharmacokinetics, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Škarydová L, Skarka A, Novotná R, Živná L, Martin HJ, Wsól V, Maser E. Partial purification and characterization of a new human membrane-bound carbonyl reductase playing a role in the deactivation of the anticancer drug oracin. Toxicology 2009; 264:52-60. [DOI: 10.1016/j.tox.2009.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 07/17/2009] [Accepted: 07/17/2009] [Indexed: 01/22/2023]
|
36
|
Identification of the promoter of human carbonyl reductase 3 (CBR3) and impact of common promoter polymorphisms on hepatic CBR3 mRNA expression. Pharm Res 2009; 26:2209-15. [PMID: 19590938 DOI: 10.1007/s11095-009-9936-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 06/29/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE Recent studies suggest that polymorphisms in human carbonyl reductase 3 (CBR3) influence the pharmacodynamics of doxorubicin. First, we sought to identify the promoter of CBR3. Next, we examined whether two CBR3 promoter polymorphisms (CBR3 -725T>C and CBR3 -326T>A) dictate promoter activity and hepatic CBR3 mRNA levels. METHODS The promoter activities of CBR3 reporter constructs were investigated in HepG2 and MCF-7 cells. CBR3 mRNA levels were documented in 95 liver samples from white (n = 62) and black (n = 33) donors. Genotype-phenotype correlation analyses were used to determine the impact of the CBR3 -725T>C and CBR3 -326T>A polymorphisms on hepatic CBR3 mRNA levels. RESULTS We identified the promoter of human CBR3. Liver samples from black donors showed higher relative CBR3 mRNA levels than samples from whites (CBR3 mRNA(blacks) = 3.0 +/- 3.1 relative fold vs. CBR3 mRNA(whites) = 1.6 +/- 1.5 relative fold, p = 0.021). The variant -725C and -326A alleles did not modify the gene reporter activities of engineered CBR3 promoter constructs. In line, hepatic CBR3 mRNA levels were not associated with CBR3 -725T>C and CBR3 -326T>A genotype status. CONCLUSIONS These studies provide the first insights into the regulation and variable hepatic expression of polymorphic CBR3.
Collapse
|
37
|
Gonzalez-Covarrubias V, Zhang J, Kalabus JL, Relling MV, Blanco JG. Pharmacogenetics of human carbonyl reductase 1 (CBR1) in livers from black and white donors. Drug Metab Dispos 2009; 37:400-7. [PMID: 19022938 PMCID: PMC2680526 DOI: 10.1124/dmd.108.024547] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 11/18/2008] [Indexed: 11/22/2022] Open
Abstract
Carbonyl reductase 1 (CBR1) reduces the anticancer drug doxorubicin into the cardiotoxic metabolite doxorubicinol. We documented the hepatic expression of CBR1 in samples from white and black donors. Concordance between ethnicity and geographical ancestry was examined with ancestry informative markers. Livers from blacks and whites showed similar CBR1 mRNA levels (CBR1 mRNA(blacks) = 4.8 +/- 4.3 relative -fold versus CBR1 mRNA(whites) = 3.6 +/- 3.6 relative -fold; p = 0.217). CBR1 protein levels did not differ between both groups (CBR1(blacks) = 8.0 +/- 3.4 nmol/g cytosolic protein versus CBR1(whites) = 9.0 +/- 4.6 nmol/g cytosolic protein; p = 0.347). The CBR1 3'-untranslated region polymorphism 1096G>A was detected in DNA samples from whites (p = 0.875; q = 0.125), and livers with homozygous G/G genotypes showed a trend toward higher CBR1 mRNA levels compared with samples with heterozygous G/A genotypes [CBR1 1096G>A((G/G)) = 4.1 +/- 4.1 relative -fold versus CBR1 1096G>A((G/A)) = 3.0 +/- 2.5 relative-fold; p = 0.266]. CBR1 1096G>A genotype status was associated with CBR1 protein levels (p = 0.030) and CBR activity expressed as the rate of synthesis of doxorubicinol (p = 0.028). Our findings warrant further studies to evaluate the impact of CBR1 1096G>A genotype status on the variable pharmacodynamics of anthracycline drugs.
Collapse
Affiliation(s)
- Vanessa Gonzalez-Covarrubias
- Department of Pharmaceutical Sciences, The State University of New York at Buffalo, 545 Cooke Hall, Buffalo, NY 14260-1200, USA
| | | | | | | | | |
Collapse
|
38
|
Studies of the metabolic stability in cells of 5-(trifluoroacetyl)thiophene-2-carboxamides and identification of more stable class II histone deacetylase (HDAC) inhibitors. Bioorg Med Chem Lett 2008; 18:6078-82. [DOI: 10.1016/j.bmcl.2008.10.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 10/07/2008] [Accepted: 10/07/2008] [Indexed: 11/19/2022]
|
39
|
Imamura Y. [Structure and function of peroxisomal tetrameric carbonyl reductase]. YAKUGAKU ZASSHI 2008; 128:1665-72. [PMID: 18981702 DOI: 10.1248/yakushi.128.1665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this paper, the structure and function of a new tetrameric carbonyl reductase (TCR) is reviewed. TCRs were purified from rabbit and pig heart, using 4-benzoylpyridine as a substrate. Partial peptide sequencing and cDNA cloning of rabbit and pig TCRs revealed that both enzymes belonged to the short-chain dehydrogenase/reductase family and that their subunits consisted of 260 amino acid residues. Rabbit and pig TCRs catalyzed the reduction of alkyl phenyl ketones, alpha-dicarbonyl compounds, quinones and retinals. Both enzymes were potently inhibited by flavonoids and fatty acids. 9,10-Phenanthrenequinone, which is efficiently reduced by rabbit and pig TCRs, mediated the formation of superoxide radical through its redox cycling in pig heart. The C-terminal sequences of rabbit and pig TCRs comprised a type 1 peroxisomal targeting signal (PTS1) Ser-Arg-Leu, suggesting that the enzymes are localized in the peroxisome. In fact, pig TCR was targeted into the peroxisomal matrix, in the case of transfection of HeLa cells with vectors expressing the enzyme. However, when the recombinant pig TCR was directly introduced into HeLa cells, the enzyme was not targeted into the peroxisomal matrix. The crystal structure of recombinant pig TCR demonstrated that the C-terminal PTS1 of each subunit of the enzyme was buried in the interior of the tetrameric molecule. These findings indicate that pig TCR is imported into the peroxisome as a monomer and then forms an active tetramer within this organelle.
Collapse
Affiliation(s)
- Yorishige Imamura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Japan.
| |
Collapse
|
40
|
Hoffmann F, Maser E. Carbonyl Reductases and Pluripotent Hydroxysteroid Dehydrogenases of the Short-chain Dehydrogenase/reductase Superfamily. Drug Metab Rev 2008; 39:87-144. [PMID: 17364882 DOI: 10.1080/03602530600969440] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Carbonyl reduction of aldehydes, ketones, and quinones to their corresponding hydroxy derivatives plays an important role in the phase I metabolism of many endogenous (biogenic aldehydes, steroids, prostaglandins, reactive lipid peroxidation products) and xenobiotic (pharmacologic drugs, carcinogens, toxicants) compounds. Carbonyl-reducing enzymes are grouped into two large protein superfamilies: the aldo-keto reductases (AKR) and the short-chain dehydrogenases/reductases (SDR). Whereas aldehyde reductase and aldose reductase are AKRs, several forms of carbonyl reductase belong to the SDRs. In addition, there exist a variety of pluripotent hydroxysteroid dehydrogenases (HSDs) of both superfamilies that specifically catalyze the oxidoreduction at different positions of the steroid nucleus and also catalyze, rather nonspecifically, the reductive metabolism of a great number of nonsteroidal carbonyl compounds. The present review summarizes recent findings on carbonyl reductases and pluripotent HSDs of the SDR protein superfamily.
Collapse
Affiliation(s)
- Frank Hoffmann
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Campus Kiel, Brunswiker Strasse, Kiel, 10, 24105, Germany
| | | |
Collapse
|
41
|
Imamura Y, Narumi R, Shimada H. Inhibition of carbonyl reductase activity in pig heart by alkyl phenyl ketones. J Enzyme Inhib Med Chem 2008; 22:105-9. [PMID: 17373555 DOI: 10.1080/14756360600954023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The inhibitory effects of alkyl phenyl ketones on carbonyl reductase activity were examined in pig heart. In this study, carbonyl reductase activity was estimated as the ability to reduce 4-benzoylpyridine to S(-)-alpha-phenyl-4-pyridylmethanol in the cytosolic fraction from pig heart (pig heart cytosol). The order of their inhibitory potencies was hexanophenone > valerophenone > heptanophenone > butyrophenone > propiophenone. The inhibitory potencies of acetophenone and nonanophenone were much lower. A significant relationship was observed between Vmax/Km values for the reduction of alkyl phenyl ketones and their inhibitory potencies for carbonyl reductase activity in pig heart cytosol. Furthermore, hexanophenone was a competitive inhibitor for the enzyme activity. These results indicate that several alkyl phenyl ketones including hexanophenone inhibit carbonyl reductase activity in pig heart cytosol, by acting as substrate inhibitors.
Collapse
Affiliation(s)
- Yorishige Imamura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1, Oe-honmachi, Kumamoto 862-0973, Japan.
| | | | | |
Collapse
|
42
|
Kassner N, Huse K, Martin HJ, Gödtel-Armbrust U, Metzger A, Meineke I, Brockmöller J, Klein K, Zanger UM, Maser E, Wojnowski L. Carbonyl reductase 1 is a predominant doxorubicin reductase in the human liver. Drug Metab Dispos 2008; 36:2113-20. [PMID: 18635746 DOI: 10.1124/dmd.108.022251] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
A first step in the enzymatic disposition of the antineoplastic drug doxorubicin (DOX) is the reduction to doxorubicinol (DOX-OL). Because DOX-OL is less antineoplastic but more cardiotoxic than the parent compound, the individual rate of this reaction may affect the antitumor effect and the risk of DOX-induced heart failure. Using purified enzymes and human tissues we determined enzymes generating DOX-OL and interindividual differences in their activities. Human tissues express at least two DOX-reducing enzymes. High-clearance organs (kidney, liver, and the gastrointestinal tract) express an enzyme with an apparent Km of approximately 140 microM. Of six enzymes found to reduce DOX, Km values in this range are exhibited by carbonyl reductase 1 (CBR1) and aldo-keto reductase (AKR) 1C3. CBR1 is expressed in these three organs at higher levels than AKR1C3, whereas AKR1C3 has higher catalytic efficiency. However, inhibition constants for DOX reduction with 4-amino-1-tert-butyl-3-(2-hydroxyphenyl)pyrazolo[3,4-d]pyrimidine (an inhibitor that can discriminate between CBR1 and AKR1C3) were identical for CBR1 and human liver cytosol, but not for AKR1C3. These results suggest that CBR1 is a predominant hepatic DOX reductase. In cytosols from 80 human livers, the expression level of CBR1 and the activity of DOX reduction varied >70- and 22-fold, respectively, but showed no association with CBR1 gene variants found in these samples. Instead, the interindividual differences in CBR1 expression and activity may be mediated by environmental factors acting via recently identified xenobiotic response elements in the CBR1 promoter. The variability in the CBR1 expression may affect outcomes of therapies with DOX, as well as with other CBR1 substrates.
Collapse
Affiliation(s)
- Nina Kassner
- Department of Pharmacology, Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, D-55101 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gavelová M, Hladíková J, Vildová L, Novotná R, Vondráček J, Krčmář P, Machala M, Skálová L. Reduction of doxorubicin and oracin and induction of carbonyl reductase in human breast carcinoma MCF-7 cells. Chem Biol Interact 2008; 176:9-18. [DOI: 10.1016/j.cbi.2008.07.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 07/18/2008] [Accepted: 07/28/2008] [Indexed: 11/16/2022]
|
44
|
Kenneke JF, Mazur CS, Ritger SE, Sack TJ. Mechanistic Investigation of the Noncytochrome P450-Mediated Metabolism of Triadimefon to Triadimenol in Hepatic Microsomes. Chem Res Toxicol 2008; 21:1997-2004. [DOI: 10.1021/tx800211t] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- John F. Kenneke
- National Exposure Research Laboratory, Student Services Authority, and Senior Service America, U.S. Environmental Protection Agency, Athens, Georgia 30605
| | - Christopher S. Mazur
- National Exposure Research Laboratory, Student Services Authority, and Senior Service America, U.S. Environmental Protection Agency, Athens, Georgia 30605
| | - Susan E. Ritger
- National Exposure Research Laboratory, Student Services Authority, and Senior Service America, U.S. Environmental Protection Agency, Athens, Georgia 30605
| | - Thomas J. Sack
- National Exposure Research Laboratory, Student Services Authority, and Senior Service America, U.S. Environmental Protection Agency, Athens, Georgia 30605
| |
Collapse
|
45
|
Flavonoids as inhibitors of human carbonyl reductase 1. Chem Biol Interact 2008; 174:98-108. [DOI: 10.1016/j.cbi.2008.05.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 05/12/2008] [Accepted: 05/13/2008] [Indexed: 11/20/2022]
|
46
|
Su JGJ, Liao PJ, Huang MC, Chu WC, Lin SC, Chang YJ. Aldo-keto reductase 1C2 is essential for 1-nitropyrene's but not for benzo[a]pyrene's induction of p53 phosphorylation and apoptosis. Toxicology 2008; 244:257-70. [DOI: 10.1016/j.tox.2007.11.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 11/23/2007] [Accepted: 11/26/2007] [Indexed: 02/01/2023]
|
47
|
Mazur CS, Kenneke JF. Cross-species comparison of conazole fungicide metabolites using rat and rainbow trout (Onchorhynchus mykiss) hepatic microsomes and purified human CYP 3A4. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2008; 42:947-954. [PMID: 18323127 DOI: 10.1021/es072049b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Ecological risk assessment frequently relies on cross-species extrapolation to predict acute toxicity from chemical exposures. A major concern for environmental risk characterization is the degree of uncertainty in assessing xenobiotic biotransformation processes. Although inherently complex, metabolite identification is critical to risk assessment since the product(s) formed may pose a greater toxicological threat than the parent molecule. This issue is further complicated by differences observed in metabolic transformation pathways among species. Conazoles represent an important class of azole fungicides that are widely used in both pharmaceutical and agricultural applications. The antifungal property of conazoles occurs via complexation with the cytochrome P450 monooxygenases (CYP) responsible for mediating fungal cell wall synthesis. This mode of action has cause for concern regarding the potential adverse impact of conazoles on the broad spectrum of CYP-based processes within mammalian and aquatic species. In this study, in vitro metabolic profiles were determined for thirteen conazole fungicides using rat and rainbow trout (Oncorhynchus mykiss) liver microsomes and purified human CYP 3A4. Results showed that 10 out of the 13 conazoles tested demonstrated identical metabolite profiles among rat and trout microsomes, and these transformations were well conserved via both aromatic and aliphatic hydroxylation and carbonyl reduction processes. Furthermore, nearly all metabolites detected in the rat and trout microsomal assays were detected within the human CYP 3A4 assays. These results indicate a high degree of metabolic conservation among species with an equivalent isozyme activity of human CYP 3A4 being present in both the rat and trout, and provides insight into xenobiotic biotransformations needed for accurate risk assessment.
Collapse
Affiliation(s)
- Christopher S Mazur
- U.S. EPA, National Exposure Research Laboratory, Ecosystems Research Division, 960 College Station Rd., Athens, GA 30605, USA.
| | | |
Collapse
|
48
|
Barski OA, Tipparaju SM, Bhatnagar A. The aldo-keto reductase superfamily and its role in drug metabolism and detoxification. Drug Metab Rev 2008; 40:553-624. [PMID: 18949601 PMCID: PMC2663408 DOI: 10.1080/03602530802431439] [Citation(s) in RCA: 381] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aldo-keto reductase (AKR) superfamily comprises enzymes that catalyze redox transformations involved in biosynthesis, intermediary metabolism, and detoxification. Substrates of AKRs include glucose, steroids, glycosylation end-products, lipid peroxidation products, and environmental pollutants. These proteins adopt a (beta/alpha)(8) barrel structural motif interrupted by a number of extraneous loops and helixes that vary between proteins and bring structural identity to individual families. The human AKR family differs from the rodent families. Due to their broad substrate specificity, AKRs play an important role in the phase II detoxification of a large number of pharmaceuticals, drugs, and xenobiotics.
Collapse
Affiliation(s)
- Oleg A Barski
- Division of Cardiology, Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky 40202, USA.
| | | | | |
Collapse
|
49
|
Lakhman SS, Chen X, Gonzalez-Covarrubias V, Schuetz EG, Blanco JG. Functional characterization of the promoter of human carbonyl reductase 1 (CBR1). Role of XRE elements in mediating the induction of CBR1 by ligands of the aryl hydrocarbon receptor. Mol Pharmacol 2007; 72:734-43. [PMID: 17569794 DOI: 10.1124/mol.107.035550] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human carbonyl reductase 1 (CBR1) metabolizes a variety of substrates, including the anticancer doxorubicin and the antipsychotic haloperidol. The transcriptional regulation of CBR1 has been largely unexplored. Therefore, we first investigated the promoter activities of progressive gene-reporter constructs encompassing up to 2.4 kilobases upstream of the translation start site of CBR1. Next, we investigated whether CBR1 mRNA levels were altered in cells incubated with prototypical receptor activators (e.g., dexamethasone and rifampicin). CBR1 mRNA levels were significantly induced (5-fold) by the ligand of the aryl hydrocarbon receptor (AHR) beta-naphthoflavone. DNA sequence analysis revealed two xenobiotic response elements ((-122)XRE and (-5783)XRE) with potential regulatory functions. CBR1 promoter constructs lacking the (-122)XRE showed diminished (9-fold) promoter activity in AHR-proficient cells incubated with beta-naphthoflavone. Fusion of (-5783)XRE to the (-2485)CBR1 reporter construct enhanced its promoter activity after incubations with beta-naphthoflavone by 5-fold. Furthermore, we tested whether the potent AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced Cbr1 expression in Ahr(+/-) and Ahr(-/-) mice. TCDD induced hepatic Cbr1 mRNA (TCDD, 2-fold) and Cbr1 protein levels (TCDD, 2-fold) in Ahr(+/-) mice compared with vehicle-injected controls. In contrast, no significant Cbr1 mRNA and Cbr1 protein induction was detected in livers from Ahr(-/-) mice treated with TCDD. These studies provide the first insights on the functional characteristics of the human CBR1 gene promoter. Our data indicate that the AHR pathway contributes to the transcriptional regulation of CBR1.
Collapse
MESH Headings
- Alcohol Oxidoreductases/genetics
- Aldehyde Reductase
- Aldo-Keto Reductases
- Animals
- Base Sequence
- Binding Sites
- Breast Neoplasms/pathology
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Female
- Genes, Reporter
- Humans
- Ligands
- Liver Neoplasms/pathology
- Luciferases/metabolism
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Polychlorinated Dibenzodioxins/pharmacology
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/analysis
- Receptors, Aryl Hydrocarbon/deficiency
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Response Elements
- Sequence Analysis, DNA
- Sequence Homology, Nucleic Acid
Collapse
Affiliation(s)
- Sukhwinder S Lakhman
- Department of Pharmaceutical Sciences, the State University of New York at Buffalo, Buffalo, New York 14260-1200, USA
| | | | | | | | | |
Collapse
|
50
|
Gonzalez-Covarrubias V, Ghosh D, Lakhman SS, Pendyala L, Blanco JG. A functional genetic polymorphism on human carbonyl reductase 1 (CBR1 V88I) impacts on catalytic activity and NADPH binding affinity. Drug Metab Dispos 2007; 35:973-80. [PMID: 17344335 PMCID: PMC2442771 DOI: 10.1124/dmd.107.014779] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human carbonyl reductase 1 (CBR1) metabolizes endogenous and xenobiotic substrates such as the fever mediator, prostaglandin E2 (PGE2), and the anticancer anthracycline drug, daunorubicin. We screened 33 CBR1 full-length cDNA samples from white and black liver donors and performed database analyses to identify genetic determinants of CBR1 activity. We pinpointed a single nucleotide polymorphism on CBR1 (CBR1 V88I) that encodes for a valine-to-isoleucine substitution for further characterization. We detected the CBR1 V88I polymorphism in DNA samples from individuals with African ancestry (p = 0.986, q = 0.014). Kinetic studies revealed that the CBR1 V88 and CBR1 I88 isoforms have different maximal velocities for daunorubicin (V(max) CBR1 V88, 181 +/- 13 versus V(max) CBR1 I88, 121 +/- 12 nmol/min . mg, p < 0.05) and PGE2 (V(max) CBR1 V88, 53 +/- 7 versus V(max) CBR1 I88, 35 +/- 4 nmol/min . mg, p < 0.01). Concomitantly, CBR1 V88 produced higher levels of the cardiotoxic metabolite daunorubicinol compared with CBR1 I88 (1.7-fold, p < 0.0001). Inhibition studies demonstrated that CBR1 V88 and CBR1 I88 are distinctively inhibited by the flavonoid, rutin (IC50 CBR1 V88, 54.0 +/- 0.4 microM versus IC50 CBR1 I88, 15.0 +/- 0.1 microM, p < 0.001). Furthermore, isothermal titration calorimetry analyses together with molecular modeling studies showed that CBR1 V88I results in CBR1 isoforms with different binding affinities for the cofactor NADPH (K(d) CBR1 V88, 6.3 +/- 0.6 microM versus K(d) CBR1 I88, 3.8 +/- 0.5 microM). These studies characterize the first functional genetic determinant of CBR1 activity toward relevant physiological and pharmacological substrates.
Collapse
|