1
|
Phillips IR, Veeravalli S, Khadayate S, Shephard EA. Metabolomic and transcriptomic analyses of Fmo5-/- mice reveal roles for flavin-containing monooxygenase 5 (FMO5) in NRF2-mediated oxidative stress response, unfolded protein response, lipid homeostasis, and carbohydrate and one-carbon metabolism. PLoS One 2023; 18:e0286692. [PMID: 37267233 PMCID: PMC10237457 DOI: 10.1371/journal.pone.0286692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/20/2023] [Indexed: 06/04/2023] Open
Abstract
Flavin-containing monooxygenase 5 (FMO5) is a member of the FMO family of proteins, best known for their roles in the detoxification of foreign chemicals and, more recently, in endogenous metabolism. We have previously shown that Fmo5-/- mice display an age-related lean phenotype, with much reduced weight gain from 20 weeks of age. The phenotype is characterized by decreased fat deposition, lower plasma concentrations of glucose, insulin and cholesterol, higher glucose tolerance and insulin sensitivity, and resistance to diet-induced obesity. In the present study we report the use of metabolomic and transcriptomic analyses of livers of Fmo5-/- and wild-type mice to identify factors underlying the lean phenotype of Fmo5-/- mice and gain insights into the function of FMO5. Metabolomics was performed by the Metabolon platform, utilising ultrahigh performance liquid chromatography-tandem mass spectroscopy. Transcriptomics was performed by RNA-Seq and results analysed by DESeq2. Disruption of the Fmo5 gene has wide-ranging effects on the abundance of metabolites and expression of genes in the liver. Metabolites whose concentration differed between Fmo5-/- and wild-type mice include several saturated and monounsaturated fatty acids, complex lipids, amino acids, one-carbon intermediates and ADP-ribose. Among the genes most significantly and/or highly differentially expressed are Apoa4, Cd36, Fitm1, Hspa5, Hyou1, Ide, Me1 and Mme. The results reveal that FMO5 is involved in upregulating the NRF2-mediated oxidative stress response, the unfolded protein response and response to hypoxia and cellular stress, indicating a role for the enzyme in adaptation to oxidative and metabolic stress. FMO5 also plays a role in stimulating a wide range of metabolic pathways and processes, particularly ones involved in lipid homeostasis, the uptake and metabolism of glucose, the generation of cytosolic NADPH, and in one-carbon metabolism. The results predict that FMO5 acts by stimulating the NRF2, XBP1, PPARA and PPARG regulatory pathways, while inhibiting STAT1 and IRF7 pathways.
Collapse
Affiliation(s)
- Ian R. Phillips
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Sunil Veeravalli
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Sanjay Khadayate
- MRC London Institute of Medical Sciences (LMS), London, United Kingdom
| | - Elizabeth A. Shephard
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| |
Collapse
|
2
|
Cleary Y, Kletzl H, Grimsey P, Heinig K, Ogungbenro K, Silber Baumann HE, Frey N, Aarons L, Galetin A, Gertz M. Estimation of FMO3 Ontogeny by Mechanistic Population Pharmacokinetic Modelling of Risdiplam and Its Impact on Drug-Drug Interactions in Children. Clin Pharmacokinet 2023; 62:891-904. [PMID: 37148485 PMCID: PMC10256639 DOI: 10.1007/s40262-023-01241-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Spinal muscular atrophy (SMA) is a progressive neuromuscular disease caused by insufficient levels of survival motor neuron (SMN) protein. Risdiplam (EvrysdiTM) increases SMN protein and is approved for the treatment of SMA. Risdiplam has high oral bioavailability and is primarily eliminated through hepatic metabolism by flavin-containing monooxygenase3 (FMO3) and cytochrome P450 (CYP) 3A, by 75% and 20%, respectively. While the FMO3 ontogeny is critical input data for the prediction of risdiplam pharmacokinetics (PK) in children, it was mostly studied in vitro, and robust in vivo FMO3 ontogeny is currently lacking. We derived in vivo FMO3 ontogeny by mechanistic population PK modelling of risdiplam and investigated its impact on drug-drug interactions in children. METHODS Population and physiologically based PK (PPK and PBPK) modelling conducted during the development of risdiplam were integrated into a mechanistic PPK (Mech-PPK) model to estimate in vivo FMO3 ontogeny. A total of 10,205 risdiplam plasma concentration-time data from 525 subjects aged 2 months-61 years were included. Six different structural models were examined to describe the in vivo FMO3 ontogeny. Impact of the newly estimated FMO3 ontogeny on predictions of drug-drug interaction (DDI) in children was investigated by simulations for dual CYP3A-FMO3 substrates including risdiplam and theoretical substrates covering a range of metabolic fractions (fm) of CYP3A and FMO3 (fmCYP3A:fmFMO3 = 10%:90%, 50%:50%, 90%:10%). RESULTS All six models consistently predicted higher FMO3 expression/activity in children, reaching a maximum at the age of 2 years with an approximately threefold difference compared with adults. Different trajectories of FMO3 ontogeny in infants < 4 months of age were predicted by the six models, likely due to limited observations for this age range. Use of this in vivo FMO3 ontogeny function improved prediction of risdiplam PK in children compared to in vitro FMO3 ontogeny functions. The simulations of theoretical dual CYP3A-FMO3 substrates predicted comparable or decreased CYP3A-victim DDI propensity in children compared to adults across the range of fm values. Refinement of FMO3 ontogeny in the risdiplam model had no impact on the previously predicted low CYP3A-victim or -perpetrator DDI risk of risdiplam in children. CONCLUSION Mech-PPK modelling successfully estimated in vivo FMO3 ontogeny from risdiplam data collected from 525 subjects aged 2 months-61 years. To our knowledge, this is the first investigation of in vivo FMO3 ontogeny by population approach using comprehensive data covering a wide age range. Derivation of a robust in vivo FMO3 ontogeny function has significant implications on the prospective prediction of PK and DDI in children for other FMO3 substrates in the future, as illustrated in the current study for FMO3 and/or dual CYP3A-FMO3 substrates. CLINICAL TRIAL REGISTRY NUMBERS NCT02633709, NCT03032172, NCT02908685, NCT02913482, NCT03988907.
Collapse
Affiliation(s)
- Yumi Cleary
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.
| | - Heidemarie Kletzl
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Paul Grimsey
- Roche Pharma Research and Early Development, Roche Innovation Center, Welwyn, UK
| | - Katja Heinig
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Hanna Elisabeth Silber Baumann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Nicolas Frey
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Leon Aarons
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Michael Gertz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
3
|
Neurobiological Basis of Aversion-Resistant Ethanol Seeking in C. elegans. Metabolites 2022; 13:metabo13010062. [PMID: 36676987 PMCID: PMC9861758 DOI: 10.3390/metabo13010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Persistent alcohol seeking despite the risk of aversive consequences is a crucial characteristic of alcohol use disorders (AUDs). Therefore, an improved understanding of the molecular basis of alcohol seeking despite aversive stimuli or punishment in animal models is an important strategy to understand the mechanism that underpins the pathology of AUDs. Aversion-resistant seeking (ARS) is characterized by disruption in control of alcohol use featured by an imbalance between the urge for alcohol and the mediation of aversive stimuli. We exploited C. elegans, a genetically tractable invertebrate, as a model to elucidate genetic components related to this behavior. We assessed the seb-3 neuropeptide system and its transcriptional regulation to progress aversion-resistant ethanol seeking at the system level. Our functional genomic approach preferentially selected molecular components thought to be involved in cholesterol metabolism, and an orthogonal test defined functional roles in ARS through behavioral elucidation. Our findings suggest that fmo-2 (flavin-containing monooxygenase-2) plays a role in the progression of aversion-resistant ethanol seeking in C. elegans.
Collapse
|
4
|
Rendić SP, Crouch RD, Guengerich FP. Roles of selected non-P450 human oxidoreductase enzymes in protective and toxic effects of chemicals: review and compilation of reactions. Arch Toxicol 2022; 96:2145-2246. [PMID: 35648190 PMCID: PMC9159052 DOI: 10.1007/s00204-022-03304-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022]
Abstract
This is an overview of the metabolic reactions of drugs, natural products, physiological compounds, and other (general) chemicals catalyzed by flavin monooxygenase (FMO), monoamine oxidase (MAO), NAD(P)H quinone oxidoreductase (NQO), and molybdenum hydroxylase enzymes (aldehyde oxidase (AOX) and xanthine oxidoreductase (XOR)), including roles as substrates, inducers, and inhibitors of the enzymes. The metabolism and bioactivation of selected examples of each group (i.e., drugs, "general chemicals," natural products, and physiological compounds) are discussed. We identified a higher fraction of bioactivation reactions for FMO enzymes compared to other enzymes, predominately involving drugs and general chemicals. With MAO enzymes, physiological compounds predominate as substrates, and some products lead to unwanted side effects or illness. AOX and XOR enzymes are molybdenum hydroxylases that catalyze the oxidation of various heteroaromatic rings and aldehydes and the reduction of a number of different functional groups. While neither of these two enzymes contributes substantially to the metabolism of currently marketed drugs, AOX has become a frequently encountered route of metabolism among drug discovery programs in the past 10-15 years. XOR has even less of a role in the metabolism of clinical drugs and preclinical drug candidates than AOX, likely due to narrower substrate specificity.
Collapse
Affiliation(s)
| | - Rachel D Crouch
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, 37204, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| |
Collapse
|
5
|
Non-cytochrome P450 enzymes involved in the oxidative metabolism of xenobiotics: Focus on the regulation of gene expression and enzyme activity. Pharmacol Ther 2021; 233:108020. [PMID: 34637840 DOI: 10.1016/j.pharmthera.2021.108020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Oxidative metabolism is one of the major biotransformation reactions that regulates the exposure of xenobiotics and their metabolites in the circulatory system and local tissues and organs, and influences their efficacy and toxicity. Although cytochrome (CY)P450s play critical roles in the oxidative reaction, extensive CYP450-independent oxidative metabolism also occurs in some xenobiotics, such as aldehyde oxidase, xanthine oxidoreductase, flavin-containing monooxygenase, monoamine oxidase, alcohol dehydrogenase, or aldehyde dehydrogenase-dependent oxidative metabolism. Drugs form a large portion of xenobiotics and are the primary target of this review. The common reaction mechanisms and roles of non-CYP450 enzymes in metabolism, factors affecting the expression and activity of non-CYP450 enzymes in terms of inhibition, induction, regulation, and species differences in pharmaceutical research and development have been summarized. These non-CYP450 enzymes are detoxifying enzymes, although sometimes they mediate severe toxicity. Synthetic or natural chemicals serve as inhibitors for these non-CYP450 enzymes. However, pharmacokinetic-based drug interactions through these inhibitors have rarely been reported in vivo. Although multiple mechanisms participate in the basal expression and regulation of non-CYP450 enzymes, only a limited number of inducers upregulate their expression. Therefore, these enzymes are considered non-inducible or less inducible. Overall, this review focuses on the potential xenobiotic factors that contribute to variations in gene expression levels and the activities of non-CYP450 enzymes.
Collapse
|
6
|
Oesch F, Honarvar N, Fabian E, Berger FI, Landsiedel R. N-vinyl compounds: studies on metabolism, genotoxicity, carcinogenicity. Arch Toxicol 2021; 95:3143-3159. [PMID: 34091723 DOI: 10.1007/s00204-021-03081-5] [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: 03/02/2021] [Accepted: 05/17/2021] [Indexed: 11/29/2022]
Abstract
Several N-vinyl compounds are produced in high volumes and are widely employed in the production of copolymers and polymers used in chemical, pharmaceutical, cosmetic and food industry. Hence, information on their genotoxicity and carcinogenicity is requisite. This review presents hitherto available information on the carcinogenicity and genotoxicity of N-vinyl compounds as well as their metabolism potentially generating genotoxic and carcinogenic derivatives. The genotoxicity and carcinogenicity of the investigated N-vinyl compounds vary widely from no observed carcinogenicity tested in lifetime bioassays in two rodent species (up to very high doses) to carcinogenicity in rats at very low doses in the absence of apparent genotoxicity. Despite of the presence of the vinyl group potentially metabolized to an epoxide followed by covalent binding to DNA, genotoxicity was observed for only one of the considered N-vinyl compounds, N-vinyl carbazole. Carcinogenicity was investigated only for two, of which one, N-vinyl pyrrolidone was carcinogenic (but not genotoxic) and ranitidine was neither carcinogenic nor genotoxic. As far as investigated, neither a metabolically formed epoxide nor a therefrom derived diol has been reported for any of the considered N-vinyl compounds. It is concluded that the information collected in this review will further the understanding of the carcinogenic potentials of N-vinyl compounds and may eventually allow approaching their prediction and prevention. A suggestion how to prevent genotoxicity in designing of N-vinyl compounds is presented. However, the available information is scarce and further research especially on the metabolism of N-vinyl compounds is highly desirable.
Collapse
Affiliation(s)
- F Oesch
- Oesch-Tox Toxicological Consulting and Expert Opinions, 55263, Ingelheim, Germany
| | - N Honarvar
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen am Rhein, Germany
| | - E Fabian
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen am Rhein, Germany
| | - F I Berger
- Regulatory Toxicology of Chemicals, BASF SE, 67056, Ludwigshafen am Rhein, Germany
| | - Robert Landsiedel
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen am Rhein, Germany.
| |
Collapse
|
7
|
Huang S, Howington MB, Dobry CJ, Evans CR, Leiser SF. Flavin-Containing Monooxygenases Are Conserved Regulators of Stress Resistance and Metabolism. Front Cell Dev Biol 2021; 9:630188. [PMID: 33644069 PMCID: PMC7907451 DOI: 10.3389/fcell.2021.630188] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/15/2021] [Indexed: 01/14/2023] Open
Abstract
Flavin-Containing Monooxygenases are conserved xenobiotic-detoxifying enzymes. Recent studies have revealed endogenous functions of FMOs in regulating longevity in Caenorhabditis elegans and in regulating aspects of metabolism in mice. To explore the cellular mechanisms of FMO's endogenous function, here we demonstrate that all five functional mammalian FMOs may play similar endogenous roles to improve resistance to a wide range of toxic stresses in both kidney and liver cells. We further find that stress-activated c-Jun N-terminal kinase activity is enhanced in FMO-overexpressing cells, which may lead to increased survival under stress. Furthermore, FMO expression modulates cellular metabolic activity as measured by mitochondrial respiration, glycolysis, and metabolomics analyses. FMO expression augments mitochondrial respiration and significantly changes central carbon metabolism, including amino acid and energy metabolism pathways. Together, our findings demonstrate an important endogenous role for the FMO family in regulation of cellular stress resistance and major cellular metabolic activities including central carbon metabolism.
Collapse
Affiliation(s)
- Shijiao Huang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Marshall B. Howington
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Craig J. Dobry
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Charles R. Evans
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Scott F. Leiser
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
8
|
A combined in vitro in-silico approach to predict the oral bioavailability of borderline BCS Class II/IV weak base albendazole and its main metabolite albendazole sulfoxide. Eur J Pharm Sci 2020; 155:105552. [PMID: 32937212 DOI: 10.1016/j.ejps.2020.105552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/28/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022]
Abstract
The aim of this study was to use a combined in vitro-in silico approach to develop a physiologically based pharmacokinetic model (PBPK) that predicts the bioavailability of albendazole (ABZ), a BCS class II/IV lipophilic weak base, and simulates its main metabolite albendazole sulphoxide (ABZSO) after oral administration of the current marketed dose of 400 mg in the fasted state. In vitro data was collected from solubility and dissolution tests performed with biorelevant media and transfer tests were carried out to evaluate the supersaturation and precipitation characteristics of ABZ upon gastric emptying. These in vitro results were used as biopharmaceutical inputs together with ABZ physicochemical properties including also permeability and in vitro metabolism data and information gathered from different clinical trials reported in the literature, were used to enable PBPK models to be developed using GastroPlus™ (version 9.7). As expected for this weak base with pKa = 3.6, ABZ exhibited a pronounced pH dependent solubility, with the solubility and extent of dissolution being greater at gastric pH and dropping significantly in the intestinal environment suggesting supersaturation and precipitation upon gastric emptying, which was confirmed by the transfer model experiments. PBPK models were set up for heathy volunteers using a full PBPK modeling approach and by implementing dynamic fluid volumes in the ACAT gut physiology in GastroPlus™. When coupling in vitro data (solubility values, dissolution rate and precipitation rate constant, etc.) for ABZ and with fitted values for the Vdss and liver systemic clearance of the sulfoxide metabolite to the PBPK model, the simulated profiles successfully predicated plasma concentrations of ABZ at 400 mg dose and simulated ABZSO at different ABZ dose levels and with different study populations, indicating the usefulness of combing in vitro biorelevant tools with PBPK modeling for the accurate prediction of ABZ bioavailability. The results obtained in this study also helped confirm that ABZ behaves as a BCS class IV compound.
Collapse
|
9
|
Matsumoto K, Hasegawa T, Ohara K, Kamei T, Koyanagi J, Akimoto M. Role of human flavin-containing monooxygenase (FMO) 5 in the metabolism of nabumetone: Baeyer-Villiger oxidation in the activation of the intermediate metabolite, 3-hydroxy nabumetone, to the active metabolite, 6-methoxy-2-naphthylacetic acid in vitro. Xenobiotica 2020; 51:155-166. [PMID: 33146575 DOI: 10.1080/00498254.2020.1843089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nabumetone (NAB) is a non-steroidal anti-inflammatory drug used clinically, and its biotransformation includes the major active metabolite 6-methoxy-2-naphthylacetic acid (6-MNA). One of the key intermediates between NAB and 6-MNA may be 3-hydroxy nabumetone (3-OH-NAB). The aim of the present study was to investigate the role of flavin-containing monooxygenase (FMO) isoform 5 in the formation of 6-MNA from 3-OH-NAB. To elucidate the biotransformation of 3-OH-NAB to 6-MNA, an authentic standard of 3-OH-NAB was synthesised and used as a substrate in an incubation with human liver samples or recombinant enzymes. The formation of 3-OH-NAB was observed after the incubation of NAB with various cytochrome P450 (CYP) isoforms. However, 6-MNA itself was rarely detected from NAB and 3-OH-NAB. Further experiments revealed a 6-MNA peak derived from 3-OH-NAB in human hepatocytes. 6-MNA was also detected in the extract obtained from 3-OH-NAB by a combined incubation of recombinant human FMO5 and human liver S9. We herein demonstrated that the reaction involves carbon-carbon cleavage catalyzed by the Baeyer-Villiger oxidation (BVO) of a carbonyl compound, the BVO substrate, such as a ketol, by FMO5. Further in vitro inhibition experiments showed that multiple non-CYP enzymes are involved in the formation of 6-MNA from 3-OH-NAB.
Collapse
Affiliation(s)
- Kaori Matsumoto
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Tetsuya Hasegawa
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Kosuke Ohara
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Tomoyo Kamei
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Junichi Koyanagi
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Masayuki Akimoto
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| |
Collapse
|
10
|
Catucci G, Aramini D, Sadeghi SJ, Gilardi G. Ligand stabilization and effect on unfolding by polymorphism in human flavin-containing monooxygenase 3. Int J Biol Macromol 2020; 162:1484-1493. [PMID: 32781122 DOI: 10.1016/j.ijbiomac.2020.08.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022]
Abstract
Pharmacogenomics is a powerful tool to prevent adverse reactions caused by different response of individuals to drug administration. Single nucleotide polymorphisms (SNPs) represent up to 90% of genetic variations among individuals. Drug metabolizing enzymes are highly polymorphic therefore the kinetic parameters of their catalytic reactions can be significantly influenced. This work reports on the unfolding process of a phase I drug metabolizing enzyme, human flavin-containing monooxygenase 3 (hFMO3) and its single nucleotide polymorphic variants (SNPs) V257M, E158K and E308G. Differential scanning calorimetry (DSC) indicates that the thermal denaturation of the enzyme is irreversible. The melting temperature (Tm) for the (Wild Type) WT and its polymorphic variants is found to be in a range from 46 °C to 50 °C. Also the activation energies of unfolding (Ea) show no significant differences among all proteins investigated (290-328 KJ/mol), except for the E308G variant that showed a significantly higher Ea of 412 KJ/mol. The presence of the bound NADP+ cofactor is found to stabilize all the variants by shifting the main Tm by 4-5 °C for all the proteins, exception made for E308G where no changes are observed. Isothermal titration calorimetry (ITC) was used to characterize the interaction of the protein with NADP+ in terms of dissociation constant (Kd), enthalpy (ΔH) and entropy (ΔS). Kd values of 1.6 and 0.7 μM, ΔH of -13.9 Kcal/mol and -16.8 Kcal/mol, ΔS of -20.5 cal/mol/deg, and -28.5 cal/mol/deg were found for V257M and E158K respectively. E308G was found to be unable to bind the NADP+ cofactor, a result that is in line with the Tm results. Circular dichroism also confirmed an overall lower stability of E308G, while NADP+ was found to give a strong positive shift of the Tm stabilizing the structure of E158K (46.2 to 50.6 °C). Previous data highlighted significant differences in terms of activity among the SNPs of hFMO3. In this work a minor impact of the SNPs was found on the stability of the enzyme in the ligand free form, except for E308G, whereas the binding of NADP+ reveals major differences among WT and polymorphic variants that are all measurable in terms of heat capacity, enthalpy and secondary structure content. These data provide the first direct evidence of ligand stabilization effects on hFMO3 that can explain the differences observed in catalytic efficiencies and serve as the starting point for the development of inhibitors of this enzyme.
Collapse
Affiliation(s)
- G Catucci
- Department of Life Sciences and Systems Biology, University of Torino, Italy
| | - D Aramini
- Department of Life Sciences and Systems Biology, University of Torino, Italy
| | - S J Sadeghi
- Department of Life Sciences and Systems Biology, University of Torino, Italy
| | - G Gilardi
- Department of Life Sciences and Systems Biology, University of Torino, Italy.
| |
Collapse
|
11
|
Öeren M, Walton PJ, Hunt PA, Ponting DJ, Segall MD. Predicting reactivity to drug metabolism: beyond P450s-modelling FMOs and UGTs. J Comput Aided Mol Des 2020; 35:541-555. [PMID: 32533369 DOI: 10.1007/s10822-020-00321-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/07/2020] [Indexed: 11/28/2022]
Abstract
We present a study based on density functional theory calculations to explore the rate limiting steps of product formation for oxidation by Flavin-containing Monooxygenase (FMO) and glucuronidation by the UDP-glucuronosyltransferase (UGT) family of enzymes. FMOs are responsible for the modification phase of metabolism of a wide diversity of drugs, working in conjunction with Cytochrome P450 (CYP) family of enzymes, and UGTs are the most important class of drug conjugation enzymes. Reactivity calculations are important for prediction of metabolism by CYPs and reactivity alone explains around 70-85% of the experimentally observed sites of metabolism within CYP substrates. In the current work we extend this approach to propose model systems which can be used to calculate the activation energies, i.e. reactivity, for the rate-limiting steps for both FMO oxidation and glucuronidation of potential sites of metabolism. These results are validated by comparison with the experimentally observed reaction rates and sites of metabolism, indicating that the presented models are suitable to provide the basis of a reactivity component within generalizable models to predict either FMO or UGT metabolism.
Collapse
Affiliation(s)
- Mario Öeren
- Optibrium Limited, Cambridge Innovation Park, Denny End Road, Cambridge, CB25 9PB, UK.
| | - Peter J Walton
- Optibrium Limited, Cambridge Innovation Park, Denny End Road, Cambridge, CB25 9PB, UK.,School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Peter A Hunt
- Optibrium Limited, Cambridge Innovation Park, Denny End Road, Cambridge, CB25 9PB, UK
| | - David J Ponting
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Leeds, LS11 5PS, UK
| | - Matthew D Segall
- Optibrium Limited, Cambridge Innovation Park, Denny End Road, Cambridge, CB25 9PB, UK
| |
Collapse
|
12
|
Veeravalli S, Phillips IR, Freire RT, Varshavi D, Everett JR, Shephard EA. Flavin-Containing Monooxygenase 1 Catalyzes the Production of Taurine from Hypotaurine. Drug Metab Dispos 2020; 48:378-385. [PMID: 32156684 DOI: 10.1124/dmd.119.089995] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022] Open
Abstract
Taurine is one of the most abundant amino acids in mammalian tissues. It is obtained from the diet and by de novo synthesis from cysteic acid or hypotaurine. Despite the discovery in 1954 that the oxygenation of hypotaurine produces taurine, the identification of an enzyme catalyzing this reaction has remained elusive. In large part, this is due to the incorrect assignment, in 1962, of the enzyme as an NAD-dependent hypotaurine dehydrogenase. For more than 55 years, the literature has continued to refer to this enzyme as such. Here we show, both in vivo and in vitro, that the enzyme that oxygenates hypotaurine to produce taurine is flavin-containing monooxygenase (FMO) 1. Metabolite analysis of the urine of Fmo1-null mice by 1H NMR spectroscopy revealed a buildup of hypotaurine and a deficit of taurine in comparison with the concentrations of these compounds in the urine of wild-type mice. In vitro assays confirmed that human FMO1 catalyzes the conversion of hypotaurine to taurine, utilizing either NADPH or NADH as cofactor. FMO1 has a wide substrate range and is best known as a xenobiotic- or drug-metabolizing enzyme. The identification that the endogenous molecule hypotaurine is a substrate for the FMO1-catalyzed production of taurine resolves a long-standing mystery. This finding should help establish the role FMO1 plays in a range of biologic processes in which taurine or its deficiency is implicated, including conjugation of bile acids, neurotransmitter, antioxidant and anti-inflammatory functions, and the pathogenesis of obesity and skeletal muscle disorders. SIGNIFICANCE STATEMENT: The identity of the enzyme that catalyzes the biosynthesis of taurine from hypotaurine has remained elusive. Here we show, both in vivo and in vitro, that flavin-containing monooxygenase 1 catalyzes the oxygenation of hypotaurine to produce taurine.
Collapse
Affiliation(s)
- Sunil Veeravalli
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| | - Ian R Phillips
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| | - Rafael T Freire
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| | - Dorsa Varshavi
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| | - Jeremy R Everett
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| | - Elizabeth A Shephard
- Department of Structural and Molecular Biology, University College London, London, United Kingdom (S.V., I.R.P., E.A.S.); School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.); and Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent, United Kingdom (R.T.F., D.V., J.R.E.)
| |
Collapse
|
13
|
Matsumoto K, Hasegawa T, Ohara K, Takei C, Kamei T, Koyanagi J, Takahashi T, Akimoto M. A metabolic pathway for the prodrug nabumetone to the pharmacologically active metabolite, 6-methoxy-2-naphthylacetic acid (6-MNA) by non-cytochrome P450 enzymes. Xenobiotica 2019; 50:783-792. [DOI: 10.1080/00498254.2019.1704097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Kaori Matsumoto
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Tetsuya Hasegawa
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Kosuke Ohara
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Chihiro Takei
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Tomoyo Kamei
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Junichi Koyanagi
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Tamiko Takahashi
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| | - Masayuki Akimoto
- Faculty of Pharmaceutical Sciences, Josai International University, Togane, Japan
| |
Collapse
|
14
|
Ancestral-sequence reconstruction unveils the structural basis of function in mammalian FMOs. Nat Struct Mol Biol 2019; 27:14-24. [PMID: 31873300 DOI: 10.1038/s41594-019-0347-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/01/2019] [Indexed: 02/02/2023]
Abstract
Flavin-containing monooxygenases (FMOs) are ubiquitous in all domains of life and metabolize a myriad of xenobiotics, including toxins, pesticides and drugs. However, despite their pharmacological importance, structural information remains bereft. To further our understanding behind their biochemistry and diversity, we used ancestral-sequence reconstruction, kinetic and crystallographic techniques to scrutinize three ancient mammalian FMOs: AncFMO2, AncFMO3-6 and AncFMO5. Remarkably, all AncFMOs could be crystallized and were structurally resolved between 2.7- and 3.2-Å resolution. These crystal structures depict the unprecedented topology of mammalian FMOs. Each employs extensive membrane-binding features and intricate substrate-profiling tunnel networks through a conspicuous membrane-adhering insertion. Furthermore, a glutamate-histidine switch is speculated to induce the distinctive Baeyer-Villiger oxidation activity of FMO5. The AncFMOs exhibited catalysis akin to human FMOs and, with sequence identities between 82% and 92%, represent excellent models. Our study demonstrates the power of ancestral-sequence reconstruction as a strategy for the crystallization of proteins.
Collapse
|
15
|
Abstract
Flavin-containing monooxygenases (FMOs) catalyze the oxygenation of numerous foreign chemicals. This review considers the roles of FMOs in the metabolism of endogenous substrates and in physiological processes, and focuses on FMOs of human and mouse. Tyramine, phenethylamine, trimethylamine, cysteamine, methionine, lipoic acid and lipoamide have been identified as endogenous or dietary-derived substrates of FMOs in vitro. However, with the exception of trimethylamine, the role of FMOs in the metabolism of these compounds in vivo is unclear. The use, as experimental models, of knockout-mouse lines deficient in various Fmo genes has revealed previously unsuspected roles for FMOs in endogenous metabolic processes. FMO1 has been identified as a novel regulator of energy balance that acts to promote metabolic efficiency, and also as being involved in the biosynthesis of taurine, by catalyzing the S-oxygenation of hypotaurine. FMO5 has been identified as a regulator of metabolic ageing and glucose homeostasis that apparently acts by sensing or responding to gut bacteria. Thus, FMOs do not function only as xenobiotic-metabolizing enzymes and there is a risk that exposure to drugs and environmental chemicals that are substrates or inducers of FMOs would perturb the endogenous functions of these enzymes.
Collapse
|
16
|
Phillips IR, Shephard EA. Flavin-containing monooxygenase 3 (FMO3): genetic variants and their consequences for drug metabolism and disease. Xenobiotica 2019; 50:19-33. [DOI: 10.1080/00498254.2019.1643515] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ian R. Phillips
- Research Department of Structural and Molecular Biology, University College London, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Elizabeth A. Shephard
- Research Department of Structural and Molecular Biology, University College London, London, UK
| |
Collapse
|
17
|
Catucci G, Sadeghi SJ, Gilardi G. A direct time-based ITC approach for substrate turnover measurements demonstrated on human FMO3. Chem Commun (Camb) 2019; 55:6217-6220. [PMID: 31074479 DOI: 10.1039/c9cc01356c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Transient binding events are a challenging issue in enzymology. Here we demostrate a time-based ITC approach to human flavin-containing monooxygenase 3, an important drug metabolising enzyme. We measure kinetic constants and we demonstrate how this approach can be exploited for measuring the inhibiton of the conversion of the key substrate trimethylamine into trimethylamine N-oxide.
Collapse
Affiliation(s)
- Gianluca Catucci
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, Torino, 10123, Italy.
| | | | | |
Collapse
|
18
|
Nguyen PTT, Parvez MM, Kim MJ, Yoo SE, Ahn S, Ghim JL, Shin JG. Physiologically Based Pharmacokinetic Modeling Approach to Predict Drug-Drug Interactions With Ethionamide Involving Impact of Genetic Polymorphism on FMO3. J Clin Pharmacol 2019; 59:880-889. [PMID: 30690726 DOI: 10.1002/jcph.1378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/02/2019] [Indexed: 11/06/2022]
Abstract
The widely used second-line antituberculosis drug ethionamide shows wide interindividual variability in its disposition; however, the relevant factors affecting this phenomenon have not been characterized. We previously reported the major contribution of flavin-containing monooxygenase 3 (FMO3) in the reductive elimination pathway of ethionamide. In this study, ethionamide metabolism was potentially inhibited by methimazole in vitro. The drug-drug interaction leading to methimazole affecting the disposition of ethionamide mediated by FMO3 was then quantitated using a bottom-up approach with a physiologically based pharmacokinetic framework. The maximum concentration (Cmax ) and area under the curve (AUC) of ethionamide were estimated to increase by 13% and 16%, respectively, when coadministered with methimazole. Subsequently, we explored the effect of FMO3 genetic polymorphism on metabolic capacity, by constructing 2 common functional variants, Lys158 -FMO3 and Gly308 -FMO3. Compared to the wild type, recombinant Lys158 -FMO3 and Gly308 -FMO3 variants significantly decreased the intrinsic clearance of ethionamide by 2% and 24%, respectively. Two prevalent functional variants of FMO3 were predicted to affect ethionamide disposition, with mean ratios of Cmax and AUC of up to 1.5 and 1.7, respectively, in comparison with the wild type. In comparing single ethionamide administration with the wild type, simulations of the combined effects of comedications and FMO3 genetic polymorphism estimated that the Cmax and AUC ratios of ethionamide increased up to 1.7 and 2.0, respectively. These findings suggested that FMO3-mediated drug-drug interaction and genetic polymorphism could be important determinants of interindividual heterogeneity in ethionamide disposition that need to be considered comprehensively to optimize the personalized dosing of ethionamide.
Collapse
Affiliation(s)
- Phuong Thi Thu Nguyen
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea.,Faculty of Pharmacy, Hai Phong University of Medicine and Pharmacy, Hai Phong, Vietnam
| | - Md Masud Parvez
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Min Jung Kim
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Sung Eun Yoo
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Sangzin Ahn
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Jong-Lyul Ghim
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and Clinical Pharmacology, PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| |
Collapse
|
19
|
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
|
20
|
Reddy VP, Jones BC, Colclough N, Srivastava A, Wilson J, Li D. An Investigation into the Prediction of the Plasma Concentration-Time Profile and Its Interindividual Variability for a Range of Flavin-Containing Monooxygenase Substrates Using a Physiologically Based Pharmacokinetic Modeling Approach. Drug Metab Dispos 2018; 46:1259-1267. [PMID: 29895591 DOI: 10.1124/dmd.118.080648] [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: 01/25/2018] [Accepted: 06/07/2018] [Indexed: 11/22/2022] Open
Abstract
Our recent paper demonstrated the ability to predict in vivo clearance of flavin-containing monooxygenase (FMO) drug substrates using in vitro human hepatocyte and human liver microsomal intrinsic clearance with standard scaling approaches. In this paper, we apply a physiologically based pharmacokinetic (PBPK) modeling and simulation approach (M&S) to predict the clearance, area under the curve (AUC), and Cmax values together with the plasma profile of a range of drugs from the original study. The human physiologic parameters for FMO, such as enzyme abundance in liver, kidney, and gut, were derived from in vitro data and clinical pharmacogenetics studies. The drugs investigated include itopride, benzydamine, tozasertib, tamoxifen, moclobemide, imipramine, clozapine, ranitidine, and olanzapine. The fraction metabolized by FMO for these drugs ranged from 21% to 96%. The developed PBPK models were verified with data from multiple clinical studies. An attempt was made to estimate the scaling factor for recombinant FMO (rFMO) using a parameter estimation approach and automated sensitivity analysis within the PBPK platform. Simulated oral clearance using in vitro hepatocyte data and associated extrahepatic FMO data predicts the observed in vivo plasma concentration profile reasonably well and predicts the AUC for all of the FMO substrates within 2-fold of the observed clinical data; seven of the nine compounds fell within 2-fold when human liver microsomal data were used. rFMO overpredicted the AUC by approximately 2.5-fold for three of the nine compounds. Applying a calculated intersystem extrapolation scalar or tissue-specific scalar for the rFMO data resulted in better prediction of clinical data. The PBPK M&S results from this study demonstrate that human hepatocytes and human liver microsomes can be used along with our standard scaling approaches to predict human in vivo pharmacokinetic parameters for FMO substrates.
Collapse
Affiliation(s)
- Venkatesh Pilla Reddy
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| | - Barry C Jones
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| | - Nicola Colclough
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| | - Abhishek Srivastava
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| | - Joanne Wilson
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| | - Danxi Li
- Departments of Modelling and Simulation, Oncology Drug Metabolism and Pharmacokinetics (V.P.R.), Departments of Drug Metabolism and Pharmacokinetics and Oncology (B.C.J., N.C., J.W.), and Department of Drug Safety and Metabolism (A.S.), IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom; and Pharmaron, Beijing, China (D.L.)
| |
Collapse
|
21
|
Nguyen PTT, Parvez MM, Kim MJ, Ho Lee J, Ahn S, Ghim JL, Shin JG. Development of a Physiologically Based Pharmacokinetic Model of Ethionamide in the Pediatric Population by Integrating Flavin-Containing Monooxygenase 3 Maturational Changes Over Time. J Clin Pharmacol 2018; 58:1347-1360. [PMID: 29878384 DOI: 10.1002/jcph.1133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 11/06/2022]
Abstract
Currently, ethionamide is the most frequently prescribed second-line antituberculosis drug in children. After extensive metabolism by flavin-containing monooxygenase (FMO) isoform 3 in the liver, the drug may exert cytotoxic effects. The comparison of children in different age groups revealed a significant age-related increase in ethionamide elimination in vivo. However, to date, the exact mechanism underlying this dynamic increase in ethionamide elimination has not been elucidated. We hypothesized that the age-dependent changes in ethionamide elimination were predominantly a result of the progressive increases in the expression and metabolic capacity of FMO3 during childhood. To test this hypothesis, a full physiologically based pharmacokinetic (PBPK) model of ethionamide was established and validated in adults through incorporation of comprehensive metabolism and transporter profiles, then expanded to the pediatric population through integration of FMO3 maturational changes over time. Thus, a good prediction PBPK model was validated successfully both in adults and children and applied to demonstrate the critical contribution of FMO3 in the mechanistic elimination pathway of ethionamide. In addition, a significant correlation between clearance and age was observed in children by accounting for ethionamide maturation, which could offer a mechanistic understanding of the age-associated changes in ethionamide elimination. In conclusion, this study underlines the benefits of in vitro-in vivo extrapolation and a quantitative PBPK approach for the investigation of transporter-enzyme interplay in ethionamide disposition and the demonstration of FMO3 ontogeny in children.
Collapse
Affiliation(s)
- Phuong Thi Thu Nguyen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Faculty of Pharmacy, Hai Phong University of Medicine and Pharmacy, Haiphong, Vietnam
| | - Md Masud Parvez
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Min Jung Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jung Ho Lee
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Sangzin Ahn
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jong-Lyul Ghim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| |
Collapse
|
22
|
Zhang T, Yang P, Wei J, Li W, Zhong J, Chen H, Cao J. Overexpression of flavin-containing monooxygenase 5 predicts poor prognosis in patients with colorectal cancer. Oncol Lett 2018; 15:3923-3927. [PMID: 29456741 DOI: 10.3892/ol.2018.7724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the expression and clinical significance of flavin-containing monooxygenase 5 (FMO5) in colorectal cancer (CRC). The expression of FMO5 was detected by immunohistochemistry in 208 colon cancer tissues and 8 normal colon tissues. Then, the correlations of FMO5 expression with several clinicopathological features were evaluated. FMO5 mRNA expression from The Cancer Genome Atlas dataset was assessed for further validation. In addition, the association of the expression of FMO5 with prognosis was further evaluated by Kaplan-Meier survival curves and Cox proportional hazards model. The FMO5 protein level in colon cancer tissues was significantly higher than that in normal colon tissues (P<0.001). Overexpression of FMO5 was associated with an advanced clinical stage of cancer (P=0.018) and lymph node metastasis (P=0.03). The TCGA dataset also demonstrated that FMO5 was upregulated in CRC with advanced clinical stage (P=0.047), lymph node metastasis (P=0.045) and distant metastasis (P=0.030). The Kaplan-Meier survival curves showed that higher FMO5 mRNA indicated a shorter overall survival in patients with CRC compared with a low expression of FMO5 (P=0.029). Cox proportional hazards regression revealed that a high FMO5 mRNA level served as an independent prognostic factor for patients with CRC (hazard ratio, 2.865; 95% confidence interval, 1.116-7.355; P=0.029). A high expression of FMO5 may serve roles in colorectal carcinogenesis and distant metastasis. FMO5 may be an independent predictive factor for the prognosis of CRC.
Collapse
Affiliation(s)
- Tong Zhang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Ping Yang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Jianchang Wei
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Wanglin Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Junbin Zhong
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Huacui Chen
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
23
|
Veeravalli S, Karu K, Scott F, Fennema D, Phillips IR, Shephard EA. Effect of Flavin-Containing Monooxygenase Genotype, Mouse Strain, and Gender on Trimethylamine N-oxide Production, Plasma Cholesterol Concentration, and an Index of Atherosclerosis. Drug Metab Dispos 2018; 46:20-25. [PMID: 29070510 PMCID: PMC5733448 DOI: 10.1124/dmd.117.077636] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/19/2017] [Indexed: 11/22/2022] Open
Abstract
The objectives of the study were to determine the contribution, in mice, of members of the flavin-containing monooxygenase (FMO) family to the production of trimethylamine (TMA) N-oxide (TMAO), a potential proatherogenic molecule, and whether under normal dietary conditions differences in TMAO production were associated with changes in plasma cholesterol concentration or with an index of atherosclerosis (Als). Concentrations of urinary TMA and TMAO and plasma cholesterol were measured in 10-week-old male and female C57BL/6J and CD-1 mice and in mouse lines deficient in various Fmo genes (Fmo1-/- , 2-/- , 4-/- , and Fmo5-/- ). In female mice most TMA N-oxygenation was catalyzed by FMO3, but in both genders 11%-12% of TMA was converted to TMAO by FMO1. Gender-, Fmo genotype-, and strain-related differences in TMAO production were accompanied by opposite effects on plasma cholesterol concentration. Plasma cholesterol was negatively, but weakly, correlated with TMAO production and urinary TMAO concentration. Fmo genotype had no effect on Als. There was no correlation between Als and either TMAO production or urinary TMAO concentration. Our results indicate that under normal dietary conditions TMAO does not increase plasma cholesterol or act as a proatherogenic molecule.
Collapse
Affiliation(s)
- Sunil Veeravalli
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Kersti Karu
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Flora Scott
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Diede Fennema
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Ian R Phillips
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| |
Collapse
|
24
|
Xu M, Bhatt DK, Yeung CK, Claw KG, Chaudhry AS, Gaedigk A, Pearce RE, Broeckel U, Gaedigk R, Nickerson DA, Schuetz E, Rettie AE, Leeder JS, Thummel KE, Prasad B. Genetic and Nongenetic Factors Associated with Protein Abundance of Flavin-Containing Monooxygenase 3 in Human Liver. J Pharmacol Exp Ther 2017; 363:265-274. [PMID: 28819071 PMCID: PMC5697103 DOI: 10.1124/jpet.117.243113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/14/2017] [Indexed: 01/20/2023] Open
Abstract
Hepatic flavin-containing mono-oxygenase 3 (FMO3) metabolizes a broad array of nucleophilic heteroatom (e.g., N or S)-containing xenobiotics (e.g., amphetamine, sulindac, benzydamine, ranitidine, tamoxifen, nicotine, and ethionamide), as well as endogenous compounds (e.g., catecholamine and trimethylamine). To predict the effect of genetic and nongenetic factors on the hepatic metabolism of FMO3 substrates, we quantified FMO3 protein abundance in human liver microsomes (HLMs; n = 445) by liquid chromatography-tandem mass chromatography proteomics. Genotyping/gene resequencing, mRNA expression, and functional activity (with benzydamine as probe substrate) of FMO3 were also evaluated. FMO3 abundance increased 2.2-fold (13.0 ± 11.4 pmol/mg protein vs. 28.0 ± 11.8 pmol/mg protein) from neonates to adults. After 6 years of age, no significant difference in FMO3 abundance was found between children and adults. Female donors exhibited modestly higher mRNA fragments per kilobase per million reads values (139.9 ± 76.9 vs. 105.1 ± 73.1; P < 0.001) and protein FMO3 abundance (26.7 ± 12.0 pmol/mg protein vs. 24.1 ± 12.1 pmol/mg protein; P < 0.05) compared with males. Six single nucleotide polymorphisms (SNPs), including rs2064074, rs28363536, rs2266782 (E158K), rs909530 (N285N), rs2266780 (E308G), and rs909531, were associated with significantly decreased protein abundance. FMO3 abundance in individuals homozygous and heterozygous for haplotype 3 (H3), representing variant alleles for all these SNPs (except rs2066534), were 50.8% (P < 0.001) and 79.5% (P < 0.01), respectively, of those with the reference homozygous haplotype (H1, representing wild-type). In summary, FMO3 protein abundance is significantly associated with age, gender, and genotype. These data are important in predicting FMO3-mediated heteroatom-oxidation of xenobiotics and endogenous biomolecules in the human liver.
Collapse
Affiliation(s)
- Meijuan Xu
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Deepak Kumar Bhatt
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Catherine K Yeung
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Katrina G Claw
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Amarjit S Chaudhry
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Andrea Gaedigk
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Robin E Pearce
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Ulrich Broeckel
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Roger Gaedigk
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Deborah A Nickerson
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Erin Schuetz
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Allan E Rettie
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - J Steven Leeder
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Kenneth E Thummel
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| | - Bhagwat Prasad
- Departments of Pharmaceutics (M.X., D.K.B., K.G.C., K.E.T., B.P.), Medicinal Chemistry (C.K.Y., A.E.R.), and Genome Sciences (D.N.), University of Washington, Seattle, Washington; Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (M.X.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.S.); Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (A.G., R.E.P., R.G., J.S.L.); and Section of Genomic Pediatrics, Department of Pediatrics, and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin (U.B.)
| |
Collapse
|
25
|
Scott F, Gonzalez Malagon SG, O'Brien BA, Fennema D, Veeravalli S, Coveney CR, Phillips IR, Shephard EA. Identification of Flavin-Containing Monooxygenase 5 (FMO5) as a Regulator of Glucose Homeostasis and a Potential Sensor of Gut Bacteria. Drug Metab Dispos 2017; 45:982-989. [PMID: 28646079 PMCID: PMC5539585 DOI: 10.1124/dmd.117.076612] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/13/2017] [Indexed: 11/24/2022] Open
Abstract
We have previously identified flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic aging. The aim of the present study was to investigate the role of FMO5 in glucose homeostasis and the impact of diet and gut flora on the phenotype of mice in which the Fmo5 gene has been disrupted (Fmo5-/- mice). In comparison with wild-type (WT) counterparts, Fmo5-/- mice are resistant to age-related changes in glucose homeostasis and maintain the higher glucose tolerance and insulin sensitivity characteristic of young animals. When fed a high-fat diet, they are protected against weight gain and reduction of insulin sensitivity. The phenotype of Fmo5-/- mice is independent of diet and the gut microbiome and is determined solely by the host genotype. Fmo5-/- mice have metabolic characteristics similar to those of germ-free mice, indicating that FMO5 plays a role in sensing or responding to gut bacteria. In WT mice, FMO5 is present in the mucosal epithelium of the gastrointestinal tract where it is induced in response to a high-fat diet. In comparison with WT mice, Fmo5-/- mice have fewer colonic goblet cells, and they differ in the production of the colonic hormone resistin-like molecule βFmo5-/- mice have lower concentrations of tumor necrosis factor α in plasma and of complement component 3 in epididymal white adipose tissue, indicative of improved inflammatory tone. Our results implicate FMO5 as a regulator of body weight and of glucose disposal and insulin sensitivity and, thus, identify FMO5 as a potential novel therapeutic target for obesity and insulin resistance.
Collapse
Affiliation(s)
- Flora Scott
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sandra G Gonzalez Malagon
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Brett A O'Brien
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Diede Fennema
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sunil Veeravalli
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Clarissa R Coveney
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Ian R Phillips
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| |
Collapse
|
26
|
Zhou W, Humphries H, Neuhoff S, Gardner I, Masson E, Al-Huniti N, Zhou D. Development of a physiologically based pharmacokinetic model to predict the effects of flavin-containing monooxygenase 3 (FMO3) polymorphisms on itopride exposure. Biopharm Drug Dispos 2017; 38:389-393. [DOI: 10.1002/bdd.2074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/27/2017] [Accepted: 02/28/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Wangda Zhou
- Quantitative Clinical Pharmacology; AstraZeneca; Waltham Massachusetts USA
| | | | | | | | - Eric Masson
- Quantitative Clinical Pharmacology; AstraZeneca; Waltham Massachusetts USA
| | - Nidal Al-Huniti
- Quantitative Clinical Pharmacology; AstraZeneca; Waltham Massachusetts USA
| | - Diansong Zhou
- Quantitative Clinical Pharmacology; AstraZeneca; Waltham Massachusetts USA
| |
Collapse
|
27
|
Phillips IR, Shephard EA. Drug metabolism by flavin-containing monooxygenases of human and mouse. Expert Opin Drug Metab Toxicol 2016; 13:167-181. [PMID: 27678284 DOI: 10.1080/17425255.2017.1239718] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Flavin-containing monooxygenases (FMOs) play an important role in drug metabolism. Areas covered: We focus on the role of FMOs in the metabolism of drugs in human and mouse. We describe FMO genes and proteins of human and mouse; the catalytic mechanism of FMOs and their significance for drug metabolism; differences between FMOs and CYPs; factors contributing to potential underestimation of the contribution of FMOs to drug metabolism; the developmental and tissue-specific expression of FMO genes and differences between human and mouse; and factors that induce or inhibit FMOs. We discuss the contribution of FMOs of human and mouse to the metabolism of drugs and how genetic variation of FMOs affects drug metabolism. Finally, we discuss the utility of animal models for FMO-mediated drug metabolism in humans. Expert opinion: The contribution of FMOs to drug metabolism may be underestimated. As FMOs are not readily induced or inhibited and their reactions are generally detoxifications, the design of drugs that are metabolized predominantly by FMOs offers clinical advantages. Fmo1(-/-),Fmo2(-/-),Fmo4(-/-) mice provide a good animal model for FMO-mediated drug metabolism in humans. Identification of roles for FMO1 and FMO5 in endogenous metabolism has implications for drug therapy and initiates an exciting area of research.
Collapse
Affiliation(s)
- Ian R Phillips
- a Institute of Structural and Molecular Biology , University College London , London , UK.,b School of Biological and Chemical Sciences , Queen Mary University of London , London , UK
| | - Elizabeth A Shephard
- a Institute of Structural and Molecular Biology , University College London , London , UK
| |
Collapse
|
28
|
Fukami T, Iida A, Konishi K, Nakajima M. Human arylacetamide deacetylase hydrolyzes ketoconazole to trigger hepatocellular toxicity. Biochem Pharmacol 2016; 116:153-61. [DOI: 10.1016/j.bcp.2016.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 07/11/2016] [Indexed: 12/20/2022]
|
29
|
Foti RS, Dalvie DK. Cytochrome P450 and Non-Cytochrome P450 Oxidative Metabolism: Contributions to the Pharmacokinetics, Safety, and Efficacy of Xenobiotics. Drug Metab Dispos 2016; 44:1229-45. [PMID: 27298339 DOI: 10.1124/dmd.116.071753] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022] Open
Abstract
The drug-metabolizing enzymes that contribute to the metabolism or bioactivation of a drug play a crucial role in defining the absorption, distribution, metabolism, and excretion properties of that drug. Although the overall effect of the cytochrome P450 (P450) family of drug-metabolizing enzymes in this capacity cannot be understated, advancements in the field of non-P450-mediated metabolism have garnered increasing attention in recent years. This is perhaps a direct result of our ability to systematically avoid P450 liabilities by introducing chemical moieties that are not susceptible to P450 metabolism but, as a result, may introduce key pharmacophores for other drug-metabolizing enzymes. Furthermore, the effects of both P450 and non-P450 metabolism at a drug's site of therapeutic action have also been subject to increased scrutiny. To this end, this Special Section on Emerging Novel Enzyme Pathways in Drug Metabolism will highlight a number of advancements that have recently been reported. The included articles support the important role of non-P450 enzymes in the clearance pathways of U.S. Food and Drug Administration-approved drugs over the past 10 years. Specific examples will detail recent reports of aldehyde oxidase, flavin-containing monooxygenase, and other non-P450 pathways that contribute to the metabolic, pharmacokinetic, or pharmacodynamic properties of xenobiotic compounds. Collectively, this series of articles provides additional support for the role of non-P450-mediated metabolic pathways that contribute to the absorption, distribution, metabolism, and excretion properties of current xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| | - Deepak K Dalvie
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| |
Collapse
|
30
|
Bohnert T, Patel A, Templeton I, Chen Y, Lu C, Lai G, Leung L, Tse S, Einolf HJ, Wang YH, Sinz M, Stearns R, Walsky R, Geng W, Sudsakorn S, Moore D, He L, Wahlstrom J, Keirns J, Narayanan R, Lang D, Yang X. Evaluation of a New Molecular Entity as a Victim of Metabolic Drug-Drug Interactions-an Industry Perspective. Drug Metab Dispos 2016; 44:1399-423. [PMID: 27052879 DOI: 10.1124/dmd.115.069096] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/31/2016] [Indexed: 12/15/2022] Open
Abstract
Under the guidance of the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), scientists from 20 pharmaceutical companies formed a Victim Drug-Drug Interactions Working Group. This working group has conducted a review of the literature and the practices of each company on the approaches to clearance pathway identification (fCL), estimation of fractional contribution of metabolizing enzyme toward metabolism (fm), along with modeling and simulation-aided strategy in predicting the victim drug-drug interaction (DDI) liability due to modulation of drug metabolizing enzymes. Presented in this perspective are the recommendations from this working group on: 1) strategic and experimental approaches to identify fCL and fm, 2) whether those assessments may be quantitative for certain enzymes (e.g., cytochrome P450, P450, and limited uridine diphosphoglucuronosyltransferase, UGT enzymes) or qualitative (for most of other drug metabolism enzymes), and the impact due to the lack of quantitative information on the latter. Multiple decision trees are presented with stepwise approaches to identify specific enzymes that are involved in the metabolism of a given drug and to aid the prediction and risk assessment of drug as a victim in DDI. Modeling and simulation approaches are also discussed to better predict DDI risk in humans. Variability and parameter sensitivity analysis were emphasized when applying modeling and simulation to capture the differences within the population used and to characterize the parameters that have the most influence on the prediction outcome.
Collapse
Affiliation(s)
- Tonika Bohnert
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Aarti Patel
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ian Templeton
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Yuan Chen
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Chuang Lu
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - George Lai
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Louis Leung
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Susanna Tse
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Heidi J Einolf
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ying-Hong Wang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Michael Sinz
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ralph Stearns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Robert Walsky
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Wanping Geng
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Sirimas Sudsakorn
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - David Moore
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ling He
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jan Wahlstrom
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jim Keirns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Rangaraj Narayanan
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Dieter Lang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Xiaoqing Yang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| |
Collapse
|
31
|
Chen Y, Zane NR, Thakker DR, Wang MZ. Quantification of Flavin-containing Monooxygenases 1, 3, and 5 in Human Liver Microsomes by UPLC-MRM-Based Targeted Quantitative Proteomics and Its Application to the Study of Ontogeny. Drug Metab Dispos 2016; 44:975-83. [PMID: 26839369 PMCID: PMC4931884 DOI: 10.1124/dmd.115.067538] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/19/2016] [Indexed: 11/22/2022] Open
Abstract
Flavin-containing monooxygenases (FMOs) have a significant role in the metabolism of small molecule pharmaceuticals. Among the five human FMOs, FMO1, FMO3, and FMO5 are the most relevant to hepatic drug metabolism. Although age-dependent hepatic protein expression, based on immunoquantification, has been reported previously for FMO1 and FMO3, there is very little information on hepatic FMO5 protein expression. To overcome the limitations of immunoquantification, an ultra-performance liquid chromatography (UPLC)-multiple reaction monitoring (MRM)-based targeted quantitative proteomic method was developed and optimized for the quantification of FMO1, FMO3, and FMO5 in human liver microsomes (HLM). A post-in silico product ion screening process was incorporated to verify LC-MRM detection of potential signature peptides before their synthesis. The developed method was validated by correlating marker substrate activity and protein expression in a panel of adult individual donor HLM (age 39-67 years). The mean (range) protein expression of FMO3 and FMO5 was 46 (26-65) pmol/mg HLM protein and 27 (11.5-49) pmol/mg HLM protein, respectively. To demonstrate quantification of FMO1, a panel of fetal individual donor HLM (gestational age 14-20 weeks) was analyzed. The mean (range) FMO1 protein expression was 7.0 (4.9-9.7) pmol/mg HLM protein. Furthermore, the ontogenetic protein expression of FMO5 was evaluated in fetal, pediatric, and adult HLM. The quantification of FMO proteins also was compared using two different calibration standards, recombinant proteins versus synthetic signature peptides, to assess the ratio between holoprotein versus total protein. In conclusion, a UPLC-MRM-based targeted quantitative proteomic method has been developed for the quantification of FMO enzymes in HLM.
Collapse
Affiliation(s)
- Yao Chen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (Y.C., M.Z.W.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina (N.R.Z., D.R.T.)
| | - Nicole R Zane
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (Y.C., M.Z.W.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina (N.R.Z., D.R.T.)
| | - Dhiren R Thakker
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (Y.C., M.Z.W.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina (N.R.Z., D.R.T.)
| | - Michael Zhuo Wang
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (Y.C., M.Z.W.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina (N.R.Z., D.R.T.)
| |
Collapse
|
32
|
Wagmann L, Meyer MR, Maurer HH. What is the contribution of human FMO3 in the N-oxygenation of selected therapeutic drugs and drugs of abuse? Toxicol Lett 2016; 258:55-70. [PMID: 27320963 DOI: 10.1016/j.toxlet.2016.06.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/12/2016] [Accepted: 06/13/2016] [Indexed: 10/21/2022]
Abstract
Little is known about the role of flavin-containing monooxygenases (FMOs) in the metabolism of xenobiotics. FMO3 is the isoform in adult human liver with the highest impact on drug metabolism. The aim of the presented study was to elucidate the contribution of human FMO3 to the N-oxygenation of selected therapeutic drugs and drugs of abuse (DOAs). Its contribution to the in vivo hepatic net clearance of the N-oxygenation products was calculated by application of an extended relative activity factor (RAF) approach to differentiate from contribution of cytochrome P450 (CYP) isoforms. FMO3 and CYP substrates were identified using pooled human liver microsomes after heat inactivation and chemical inhibition, or single enzyme incubations. Kinetic parameters were subsequently determined using recombinant human enzymes and mass spectrometric analysis via authentic reference standards or simple peak areas of the products divided by those of the internal standard. FMO3 was identified as enzyme mainly responsible for the formation of N,N-diallyltryptamine N-oxide and methamphetamine hydroxylamine (>80% contribution for both). A contribution of 50 and 30% was calculated for the formation of N,N-dimethyltryptamine N-oxide and methoxypiperamide N-oxide, respectively. However, FMO3 contributed with less than 5% to the formation of 3-bromomethcathinone hydroxylamine, amitriptyline N-oxide, and clozapine N-oxide. There was no significant difference in the contributions when using calibrations with reference metabolite standards or peak area ratio calculations. The successful application of a modified RAF approach including FMO3 proved the importance of FMO3 in the N-oxygenation of DOAs in human metabolism.
Collapse
Affiliation(s)
- Lea Wagmann
- Department of Experimental and Clinical Toxicology, Saarland University, Homburg, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Saarland University, Homburg, Germany; Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hans H Maurer
- Department of Experimental and Clinical Toxicology, Saarland University, Homburg, Germany.
| |
Collapse
|
33
|
Gonzalez Malagon SG, Melidoni AN, Hernandez D, Omar BA, Houseman L, Veeravalli S, Scott F, Varshavi D, Everett J, Tsuchiya Y, Timms JF, Phillips IR, Shephard EA. The phenotype of a knockout mouse identifies flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic ageing. Biochem Pharmacol 2015; 96:267-77. [PMID: 26049045 PMCID: PMC4509511 DOI: 10.1016/j.bcp.2015.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
Abstract
We report the production and metabolic phenotype of a mouse line in which the Fmo5 gene is disrupted. In comparison with wild-type (WT) mice, Fmo5(-/-) mice exhibit a lean phenotype, which is age-related, becoming apparent after 20 weeks of age. Despite greater food intake, Fmo5(-/-) mice weigh less, store less fat in white adipose tissue (WAT), have lower plasma glucose and cholesterol concentrations and enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure, with no increase in physical activity. An increase in respiratory exchange ratio during the dark phase, the period in which the mice are active, indicates a switch from fat to carbohydrate oxidation. In comparison with WT mice, the rate of fatty acid oxidation in Fmo5(-/-) mice is higher in WAT, which would contribute to depletion of lipid stores in this tissue, and lower in skeletal muscle. Five proteins were down regulated in the liver of Fmo5(-/-) mice: aldolase B, ketohexokinase and cytosolic glycerol 3-phosphate dehydrogenase (GPD1) are involved in glucose or fructose metabolism and GPD1 also in production of glycerol 3-phosphate, a precursor of triglyceride biosynthesis; HMG-CoA synthase 1 is involved in cholesterol biosynthesis; and malic enzyme 1 catalyzes the oxidative decarboxylation of malate to pyruvate, in the process producing NADPH for use in lipid and cholesterol biosynthesis. Down regulation of these proteins provides a potential explanation for the reduced fat deposits and lower plasma cholesterol characteristic of Fmo5(-/-) mice. Our results indicate that disruption of the Fmo5 gene slows metabolic ageing via pleiotropic effects.
Collapse
Affiliation(s)
| | - Anna N Melidoni
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Diana Hernandez
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Bilal A Omar
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Lyndsey Houseman
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Sunil Veeravalli
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Flora Scott
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Dorsa Varshavi
- Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
| | - Jeremy Everett
- Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
| | - Yugo Tsuchiya
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - John F Timms
- Women's Cancer, Institute for Women's Health, University College London, London WC1E 6BT, UK
| | - Ian R Phillips
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
34
|
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
|
35
|
Nakamaru Y, Hayashi Y, Ikegawa R, Kinoshita S, Madera BP, Gunput D, Kawaguchi A, Davies M, Mair S, Yamazaki H, Kume T, Suzuki M. Metabolism and disposition of the dipeptidyl peptidase IV inhibitor teneligliptin in humans. Xenobiotica 2013; 44:242-53. [DOI: 10.3109/00498254.2013.816891] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Effects upon in-vivo nicotine metabolism reveal functional variation in FMO3 associated with cigarette consumption. Pharmacogenet Genomics 2013; 23:62-8. [PMID: 23211429 DOI: 10.1097/fpc.0b013e32835c3b48] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Flavin-containing monooxygenases (FMO) catalyze the metabolism of nucleophilic heteroatom-containing drugs and xenobiotics, including nicotine. Rare mutations in FMO3 are responsible for defective N-oxidation of dietary trimethylamine leading to trimethylaminuria, and common genetic variation in FMO3 has been linked to interindividual variability in metabolic function that may be substrate specific. METHODS A genetic model of CYP2A6 function is used as a covariate to reveal functional polymorphism in FMO3 that indirectly influences the ratio of deuterated nicotine metabolized to cotinine following oral administration. The association is tested between FMO3 haplotype and cigarette consumption in a set of nicotine-dependent smokers. RESULTS FMO3 haplotype, based on all common coding variants in Europeans, significantly predicts nicotine metabolism and accounts for ∼2% of variance in the apparent percent of nicotine metabolized to cotinine. The metabolic ratio is not associated with FMO2 haplotype or an FMO1 expression quantitative trait locus. Cross-validation demonstrates calculated FMO3 haplotype parameters to be robust and significantly improve the predictive nicotine metabolism model over CYP2A6 genotype alone. Functional classes of FMO3 haplotypes, as determined by their influence on nicotine metabolism to cotinine, are also significantly associated with cigarettes per day in nicotine-dependent European Americans (n=1025, P=0.04), and significantly interact (P=0.016) with CYP2A6 genotype to predict cigarettes per day. CONCLUSION These findings suggest that common polymorphisms in FMO3 influence nicotine clearance and that these genetic variants in turn influence cigarette consumption.
Collapse
|
37
|
Molecular and functional characterization of flavin-containing monooxygenases in cynomolgus macaque. Biochem Pharmacol 2013; 85:1837-47. [DOI: 10.1016/j.bcp.2013.04.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/16/2013] [Accepted: 04/16/2013] [Indexed: 11/23/2022]
|
38
|
Hibar DP, Stein JL, Ryles AB, Kohannim O, Jahanshad N, Medland SE, Hansell NK, McMahon KL, de Zubicaray GI, Montgomery GW, Martin NG, Wright MJ, Saykin AJ, Jack CR, Weiner MW, Toga AW, Thompson PM. Genome-wide association identifies genetic variants associated with lentiform nucleus volume in N = 1345 young and elderly subjects. Brain Imaging Behav 2013; 7:102-15. [PMID: 22903471 PMCID: PMC3779070 DOI: 10.1007/s11682-012-9199-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Deficits in lentiform nucleus volume and morphometry are implicated in a number of genetically influenced disorders, including Parkinson's disease, schizophrenia, and ADHD. Here we performed genome-wide searches to discover common genetic variants associated with differences in lentiform nucleus volume in human populations. We assessed structural MRI scans of the brain in two large genotyped samples: the Alzheimer's Disease Neuroimaging Initiative (ADNI; N = 706) and the Queensland Twin Imaging Study (QTIM; N = 639). Statistics of association from each cohort were combined meta-analytically using a fixed-effects model to boost power and to reduce the prevalence of false positive findings. We identified a number of associations in and around the flavin-containing monooxygenase (FMO) gene cluster. The most highly associated SNP, rs1795240, was located in the FMO3 gene; after meta-analysis, it showed genome-wide significant evidence of association with lentiform nucleus volume (P MA = 4.79 × 10(-8)). This commonly-carried genetic variant accounted for 2.68 % and 0.84 % of the trait variability in the ADNI and QTIM samples, respectively, even though the QTIM sample was on average 50 years younger. Pathway enrichment analysis revealed significant contributions of this gene to the cytochrome P450 pathway, which is involved in metabolizing numerous therapeutic drugs for pain, seizures, mania, depression, anxiety, and psychosis. The genetic variants we identified provide replicated, genome-wide significant evidence for the FMO gene cluster's involvement in lentiform nucleus volume differences in human populations.
Collapse
Affiliation(s)
- Derrek P. Hibar
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - Jason L. Stein
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - April B. Ryles
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - Omid Kohannim
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - Neda Jahanshad
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - Sarah E. Medland
- Genetic Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
- Neurogenetics Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
- Broad Institute of Harvard and MIT, Boston, MA, USA
| | - Narelle K. Hansell
- Genetic Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Katie L. McMahon
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Greig I. de Zubicaray
- Functional Magnetic Resonance Imaging Laboratory, School of Psychology, University of Queensland, Brisbane, Queensland, Australia
| | - Grant W. Montgomery
- Genetic Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Nicholas G. Martin
- Genetic Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Margaret J. Wright
- Genetic Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Michael W. Weiner
- Departments of Radiology, Medicine, Psychiatry, UC San Francisco, San Francisco, CA, USA
- Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | - Paul M. Thompson
- Imaging Genetics Center at the Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Neuroscience Research Building 225E 635 Charles Young Drive, Los Angeles, CA 90095-1769, USA
| | | |
Collapse
|
39
|
|
40
|
Effect of human flavin-containing monooxygenase 3 polymorphism on the metabolism of aurora kinase inhibitors. Int J Mol Sci 2013; 14:2707-16. [PMID: 23358255 PMCID: PMC3588010 DOI: 10.3390/ijms14022707] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/22/2012] [Accepted: 01/18/2013] [Indexed: 11/17/2022] Open
Abstract
Aurora kinases were recently identified as a potential target in anticancer therapy and, amongst their available inhibitors, Tozasertib (VX-680) and Danusertib (PHA-739358) have been indicated as possible substrates of human flavin-containing monooxygenase 3 (hFMO3). Here we report the in vitro rate of oxidation of these drugs by wild-type hFMO3 and its polymorphic variant V257M. The conversion of Tozasertib and Danusertib to their corresponding metabolites, identified by LC-MS, by the purified wild-type and V257M hFMO3 show significant differences. In the case of Tozasertib, the V257M variant shows a catalytic efficiency, expressed as kcat/Km, similar to the wild-type: 0.39 ± 0.06 min−1μM−1 for V257M compared to 0.33 ± 0.04 min−1μM−1 for the wild type. On the other hand, in the case of Danusertib, V257M shows a 3.4× decrease in catalytic efficiency with kcat/Km values of 0.05 ± 0.01 min−1μM−1 for V257M and 0.17 ± 0.03 min−1μM−1 for the wild type. These data reveal how a simple V257M substitution ascribed to a single nucleotide polymorphism affects the N-oxidation of relevant anticancer drugs, with important outcome in their therapeutic effects. These findings demonstrate that codon 257 is important for activity of the hFMO3 gene and the codon change V to M has an effect on the catalytic efficiency of this enzyme.
Collapse
|
41
|
Influence of FMO1 and 3 polymorphisms on serum olanzapine and its N-oxide metabolite in psychiatric patients. THE PHARMACOGENOMICS JOURNAL 2012; 13:544-50. [DOI: 10.1038/tpj.2012.47] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/14/2012] [Accepted: 10/09/2012] [Indexed: 11/08/2022]
|
42
|
Ozhan G, Topal P, Alpertunga B. Flavin-containing monooxygenase 3 gene polymorphisms in Turkish population. Toxicol Mech Methods 2012; 22:461-5. [PMID: 22409263 DOI: 10.3109/15376516.2012.672481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Flavin-containing monooxygenases (FMOs) represent the second most important human monooxygenase system, after cytochrome P450s (CYPs) and catalyze the oxygenation of many chemicals containing nitrogen-, sulphur-, phosphorous-, selenium- and other nucleophilic heteroatoms. FMO3 is the prominent FMO form in adult human liver. For FMO3, both interindividual variability within a single ethnic group and variability between ethnic groups have been reported. In our study, three prevalent functional FMO3 variants (E158K, V257M, and E308G) were genotyped in healthy Turkish people by polymerase chain reaction (PCR)-restriction fragment length polymorphism (RFLP) methods. The frequencies of alleles and haplotypes were compared with those obtained from different populations. It was found that FMO3 158K, 257M and 308G alleles, demonstrate impaired metabolism toward many FMO3 substrates, were observed frequently in Turkish population similar to the other populations. Also, the frequencies of haplotypes were determined based on individual allelic frequencies and it was observed that the most common haplotypes were haplotip EVE and KVE (E158K/V257M/E308G), which together accounted for 80% of all haplotypes. The obtained data from the present study could be useful for further studies assessing sensitivity to therapeutic drugs, environmental toxicants and common disease.
Collapse
Affiliation(s)
- Gül Ozhan
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
| | | | | |
Collapse
|
43
|
Irving RM, Elfarra AA. Role of reactive metabolites in the circulation in extrahepatic toxicity. Expert Opin Drug Metab Toxicol 2012; 8:1157-72. [PMID: 22681489 DOI: 10.1517/17425255.2012.695347] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Reactive metabolite-mediated toxicity is frequently limited to the organ where the electrophilic metabolites are generated. Some reactive metabolites, however, might have the ability to translocate from their site of formation. This suggests that for these reactive metabolites, investigations into the role of organs other than the one directly affected could be relevant to understanding the mechanism of toxicity. AREAS COVERED The authors discuss the physiological and biochemical factors that can enable reactive metabolites to cause toxicity in an organ distal from the site of generation. Furthermore, the authors present a case study which describes studies that demonstrate that S-(1,2-dichlorovinyl)-L-cysteine sulfoxide (DCVCS) and N-acetyl-S-(1,2-dichlorovinyl-L-cysteine sulfoxide (N-AcDCVCS), reactive metabolites of the known trichloroethylene metabolites S-(1,2-dichlorovinyl)-L-cysteine (DCVC), and N-acetyl-S-(1,2-dichlorovinyl)-L-cysteine (N-AcDCVC), are generated in the liver and translocate through the circulation to the kidney to cause nephrotoxicity. EXPERT OPINION The ability of reactive metabolites to translocate could be important to consider when investigating mechanisms of toxicity. A mechanistic approach, similar to the one described for DCVCS and N-AcDCVCS, could be useful in determining the role of circulating reactive metabolites in extrahepatic toxicity of drugs and other chemicals. If this is the case, intervention strategies that would not otherwise be feasible might be effective for reducing extrahepatic toxicity.
Collapse
Affiliation(s)
- Roy M Irving
- University of Wisconsin-Madison, School of Veterinary Medicine, Department of Comparative Biosciences and Molecular and Environmental Toxicology Center, Madison, WI 53706, USA
| | | |
Collapse
|
44
|
Lai WG, Farah N, Moniz GA, Wong YN. A Baeyer-Villiger oxidation specifically catalyzed by human flavin-containing monooxygenase 5. Drug Metab Dispos 2011; 39:61-70. [PMID: 20947616 DOI: 10.1124/dmd.110.035360] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
10-((4-Hydroxypiperidin-1-yl)methyl)chromeno[4,3,2-de]phthalazin-3(2H)-one (E7016), an inhibitor of poly(ADP-ribose) polymerase, is being developed for anticancer therapy. One of the major metabolites identified in preclinical animal studies was the product of an apparent oxidation and ring opening of the 4-hydroxypiperidine. In vitro, this oxidized metabolite could not be generated by incubating E7016 with animal or human liver microsomes. Further studies revealed the formation of this unique metabolite in hepatocytes. In a NAD(P)(+)-dependent manner, this metabolite was also generated by liver S9 fractions and recombinant human flavin-containing monooxygenase (FMO) 5 that was fortified with liver cytosol fractions. In animal and human liver S9, this metabolic pathway could be inhibited by 4-methylpyrazole, bis-p-nitrophenylphosphate (BNPP), or a brief heat treatment at 50°C. Based on these results, the overall metabolic pathway was believed to involve a two-step oxidation process: dehydrogenation of the secondary alcohol in liver cytosol followed by an FMO5-mediated Baeyer-Villiger oxidation in liver microsomes. The two oxidation steps were coupled via regeneration of NAD(P)(+) and NAD(P)H. To further confirm this mechanism, the proposed ketone intermediate was independently synthesized. In an NAD(P)H-dependent manner, the synthetic ketone intermediate was metabolized to the same ring-opened metabolite in animal and human liver microsomes. This metabolic reaction was also inhibited by BNPP or a brief heat treatment at 50°C. Methimazole, the substrate/inhibitor of FMO1 and FMO3, did not inhibit this reaction. The specificity of FMO5 toward catalyzing this Baeyer-Villiger oxidation was further demonstrated by incubating the synthetic ketone intermediate in recombinant enzymes.
Collapse
Affiliation(s)
- W George Lai
- Drug Metabolism and Pharmacokinetics-Andover, Biopharmaceutical Assessments, Eisai Inc., Andover, Massachusetts 01810-2441, USA.
| | | | | | | |
Collapse
|
45
|
Reddy RR, Ralph EC, Motika MS, Zhang J, Cashman JR. Characterization of human flavin-containing monooxygenase (FMO) 3 and FMO5 expressed as maltose-binding protein fusions. Drug Metab Dispos 2010; 38:2239-45. [PMID: 20810540 PMCID: PMC2993457 DOI: 10.1124/dmd.110.033639] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 08/31/2010] [Indexed: 11/22/2022] Open
Abstract
The flavin-containing monooxygenase (FMO) family of enzymes oxygenates nucleophilic xenobiotics and endogenous substances. Human FMO3 and FMO5 are the predominant FMO forms in adult liver. These enzymes are naturally membrane-bound, and recombinant proteins are commercially available as microsomal preparations from insect cells (i.e., Supersome FMO). As an alternative, FMO3 has previously been expressed as a soluble protein, through use of an N-terminal maltose-binding protein (MBP) fusion. In the current study, MBP fusions of both human FMO3 and FMO5 were prepared to >90% purity in the presence of detergent and characterized for biochemical and kinetic parameters, and the parameters were compared with those of Supersome FMO samples. Although MBP-FMO enzymes afforded lower rates of turnover than the corresponding Supersome FMOs, both types of FMO showed identical substrate dependencies and similar responses to changes in assay conditions. Of interest, the FMO3 enzymes showed a 2-fold activation of k(cat)/K(m) in the presence of Triton X-100. Oligomeric analysis of MBP-FMO3 also showed disassociation from a high-order oligomeric form to a monomeric status in the presence of Triton X-100. This report serves as the first direct comparison between Supersome FMOs and the corresponding MBP fusions and the first report of a detergent-based activation of k(cat)/K(m) that corresponds to changes in oligomerization.
Collapse
Affiliation(s)
- Robert R Reddy
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | | | | | | | | |
Collapse
|
46
|
Narayana B, Veena K, Ashwani K, Shetty DN. New reagents for the spectrophotometric determination of ranitidine hydrochloride. ECLÉTICA QUÍMICA 2010. [DOI: 10.1590/s0100-46702010000300010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
47
|
Ochiai Y, Sakurai E, Nomura A, Itoh K, Tanaka Y. Metabolism of nicotine in rat lung microvascular endothelial cells. J Pharm Pharmacol 2010; 58:403-7. [PMID: 16536909 DOI: 10.1211/jpp.58.3.0016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
The aim of this study was to examine whether cultured rat lung microvascular endothelial cells (LMECs), which constitute the gas-blood barrier, have the ability to metabolize nicotine. Nicotine was biotransformed to cotinine and nicotine N′-oxide by cytochrome 450 (CYP) and flavin-containing monooxyganase (FMO), respectively, in rat LMECs. The intrinsic clearance (Vmax1/Km1) for the cotinine formation was about 20 times as high as that for the trans-nicotine N′-oxide formation in the low-Km phase, indicating that oxidation by CYP was much higher than that by FMO. On the other hand, as shown in Eadie-Hofstee plots, the formation of cis-nicotine N′-oxide was monophasic, whereas the plot for the trans-nicotine N′-oxide formation was clearly biphasic. These results suggest that nicotine N′-oxide was stereoselectively metabolized to cis and trans forms. However, in the high-Km phase there was no significant difference in N′-oxidation between the cis and trans forms. Moreover, we suggest that CYP2C11 and CYP3A2 are key players in the metabolism to cotinine of nicotine in rat LMECs using the respective enzyme inhibitors (tranylcypromine and troleandomycine). On the other hand, methimazole (5 μm) caused 73 and 45% decreases in the formation of N′-oxides of cis- and trans- enantiomers, respectively, demonstrating the presence of FMO in rat LMECs. These results suggest that rat LMEC enzymes can convert substrates of exogenous origin such as nicotine for detoxication, indicating LMECs are an important barrier for metabolic products, besides hepatic cells.
Collapse
Affiliation(s)
- Yoshinori Ochiai
- Department of Pharmaceutics I, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Sendai 981-8558, Japan
| | | | | | | | | |
Collapse
|
48
|
Novick RM, Vezina CM, Elfarra AA. Isoform distinct time-, dose-, and castration-dependent alterations in flavin-containing monooxygenase expression in mouse liver after 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment. Biochem Pharmacol 2009; 79:1345-51. [PMID: 20036217 DOI: 10.1016/j.bcp.2009.12.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/16/2009] [Accepted: 12/17/2009] [Indexed: 01/07/2023]
Abstract
Flavin-containing monooxygenase (FMO) expression in male mouse liver is altered after 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure or castration. Because TCDD is slowly eliminated from the body, we examined hepatic Fmo mRNA alterations for up to 32 days following 10 or 64 microg/kg TCDD exposure by oral gavage in male C57BL/6J mice. Fmo2 mRNA was significantly induced at 1, 4, and 8 days whereas Fmo3 mRNA was also induced at 32 days relative to controls. Fmo3 mRNA levels exhibited a dose-dependent increase at 4, 8, and 32 days after exposure; Fmo1, Fmo4, and Fmo5 mRNA did not exhibit clear trends. Because castration alone also increased Fmo2, Fmo3, and Fmo4 mRNA we examined the combined effects of castration and TCDD treatment on FMO expression. A greater than additive effect was observed with Fmo2 and Fmo3 mRNA expression. Fmo2 mRNA exhibited a 3-5-fold increase after castration or 10 microg/kg TCDD exposure by oral gavage, whereas an approximately 20-fold increase was observed between the sham-castrated control and castrated TCDD-treated mice. Similarly, treatment with 10 microg/kg TCDD alone increased Fmo3 mRNA 130- and 180-fold in the sham-castrated and castrated mice compared to their controls respectively, whereas, Fmo3 mRNA increased approximately 1900-fold between the sham control and castrated TCDD-treated mice. An increase in hepatic Fmo3 protein in TCDD-treated mice was observed by immunoblotting and assaying methionine S-oxidase activity. Collectively, these results provide evidence for isoform distinct time-, dose-, and castration-dependent effects of TCDD on FMO expression and suggest cross-talk between TCDD and testosterone signal transduction pathways.
Collapse
Affiliation(s)
- Rachel M Novick
- Department of Comparative Biosciences and Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | |
Collapse
|
49
|
Shimizu M. [Individual differences of drug-metabolizing enzymes as determinants for the metabolic fate of chemicals--a study of trimethylamine and flavin-containing monooxygenase 3-]. YAKUGAKU ZASSHI 2009; 129:1351-6. [PMID: 19881207 DOI: 10.1248/yakushi.129.1351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Individual differences of drug-metabolizing enzymes are important determinants for the metabolic fate of chemicals. This article focuses on polymorphic human flavin-containing monooxygenase 3 (FMO3) and dietary-derived trimethylamine. Malodorous trimethylamine is generally converted to odorless trimethylamine N-oxide by liver microsomal FMO3. Trimethylaminuria is caused by functional disorder of FMO3. In this study mutations of the FMO3 gene were examined in self-reported Japanese trimethylaminuria subjects that showed low FMO3 metabolic capacity in urine tests. Nine novel polymorphisms in the FMO3 gene were discovered in self-reported Japanese volunteers. Functional analyses of recombinant FMO3 proteins suggested that these FMO3 gene mutations were one of the causal factors for decreased FMO3 function resulting in trimethylaminuria. Inter-individual variations of FMO3-mediated microsomal oxygenation activities, levels of FMO3 protein and FMO3 mRNA, and its modification in liver microsomes from Japanese samples were observed. Both genetic polymorphisms in the 5'-upstream of the FMO3 gene and some hormonal changes related to menstruation may be causal factors for inter- and/or intra- individual expression levels of FMO3. To assess the palliative cares, it was found that absorbed levels of trimethylamine in vivo would be possibly controlled by selection of precursor foods like fish containing a variety of trimethylamine amounts. These lines of evidence suggest that individual differences of FMO3 are important determinants for the metabolic fate of dietary-derived trimethylamine.
Collapse
Affiliation(s)
- Makiko Shimizu
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan.
| |
Collapse
|
50
|
Lickteig AJ, Riley R, Melton RJ, Reitz BA, Fischer HD, Stevens JC. Expression and characterization of functional dog flavin-containing monooxygenase 3. Drug Metab Dispos 2009; 37:1987-90. [PMID: 19635782 DOI: 10.1124/dmd.109.027714] [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: 02/13/2025] Open
Abstract
Mammalian flavin-containing monooxygenase (FMO) enzymes catalyze oxidation at nucleophilic, heteroatom centers and are important for drug, xenobiotic, and endogenous substrate metabolism. In human liver, human FMO3 (hFMO3) is the most abundant FMO isoform and is known to contribute to the hepatic clearance of a variety of clinical drugs. The purpose of the current study was to express and compare the dog (beagle) FMO3 (dFMO3) to hFMO3. A full-length dFMO3 cDNA was obtained from liver by reverse transcription-polymerase chain reaction. Using a baculovirus expression system in Spodoptera frugiperda insect cells, dFMO3 was expressed to protein levels of 0.50 nmol/mg, as determined by liquid chromatography-fluorescence detection. Expressed dFMO3 displayed Michaelis-Menten kinetics, catalyzing NADPH-dependent N-oxidation of benzydamine, with K(m) and V(max) values of 18.6 microM and 0.63 nmol N-oxide formed/min/nmol of enzyme, respectively. Benzydamine N-oxidation catalyzed by hFMO3 showed values of 42.6 microM (K(m)) and 3.56 nmol/min/nmol of enzyme (V(max)). Human FMO3 was observed to catalyze the S-oxidation of sulindac sulfide, with respective K(m) and V(max) values of 69.3 microM and 35.4 nmol/min/nmol of enzyme. dFMO3 also catalyzed sulindac sulfide S-oxidation with 6.8 nmol/min/nmol of enzyme being the highest velocity observed. Finally, Western blot analysis indicated protein expression levels of dFMO3 in pooled dog liver and lung microsomes to be 27 and 9 pmol/mg, respectively. In summary, dFMO3 appears to be a functional enzyme expressed at appreciable levels in liver, but one with some kinetic properties that are substantially different from its human homolog hFMO3.
Collapse
Affiliation(s)
- Andrew J Lickteig
- Pharmacokinetics, Dynamics and Metabolism, Chesterfield, MO 63017, USA.
| | | | | | | | | | | |
Collapse
|