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
|
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: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [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
|
3
|
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
|
4
|
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 2017; 46:20-25. [PMID: 29070510 PMCID: PMC5733448 DOI: 10.1124/dmd.117.077636] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [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
|
5
|
Tanino T, Bando T, Komada A, Nojiri Y, Okada Y, Ueda Y, Sakurai E. Hepatic Flavin-Containing Monooxygenase 3 Enzyme Suppressed by Type 1 Allergy-Produced Nitric Oxide. Drug Metab Dispos 2017; 45:1189-1196. [DOI: 10.1124/dmd.117.076570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
|
6
|
Kupsco A, Schlenk D. Developmental expression and regulation of flavin-containing monooxygenase by the unfolded protein response in Japanese medaka (Oryzias latipes). Comp Biochem Physiol C Toxicol Pharmacol 2017; 191:7-13. [PMID: 27612667 DOI: 10.1016/j.cbpc.2016.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022]
Abstract
Flavin-containing monooxygenases (FMOs) play a key role in xenobiotic metabolism, are regulated by environmental conditions, and are differentially regulated during mammalian development. Japanese medaka (Oryzias latipes) are a common model organism for toxicological studies. The goal of the current research was to characterize developmental expression and regulation of FMOs in Japanese medaka embryos to better understand the role of FMOs in this model species. Five putative medaka fmos were characterized from the medaka genome through the National Center for Biotechnology Information (NCBI) database by protein motifs and alignments, then identified as fmo4, fmo5A, fmo5B, fmo5C and fmo5D for the current study. Fmo gene expression was analyzed at 1dpf, 3dpf, 6dpf and 9dpf and distinct developmental patterns of expression were observed. Fmo4 and fmo5D increased 3-fold during mid organogenesis (6dpf), while fmo5B and fmo5C decreased significantly in early organogenesis (3dpf) and fmo5A was unaltered. Promoter analysis was performed for transcription factor binding sites and indicated regulation by developmental factors and a role for the unfolded protein response in fmo modulation. Fmo regulation by the UPR was assessed with treatments of 1μg/ml, 2μg/ml, and 4μg/ml Tunicamycin (Tm), and 2mM and 4mM dithiothreitol (DTT), well-known inducers of endoplasmic reticulum stress, for 24h from 5-6dpf. High concentrations to Tm induced fmo4 and fmo5A up to two-fold, while DTT significantly decreased expression of fmo5A, fmo5B, and fmo5C. Results suggest that medaka fmos are variably regulated by the UPR during organogenesis with variable developmental expression, and suggesting potential stage-dependent activation or detoxification of xenobiotics.
Collapse
Affiliation(s)
- Allison Kupsco
- Environmental Toxicology Program, Department of Environmental Sciences, University of California-Riverside, Riverside, CA, United States.
| | - Daniel Schlenk
- Environmental Toxicology Program, Department of Environmental Sciences, University of California-Riverside, Riverside, CA, United States
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Cruciani G, Valeri A, Goracci L, Pellegrino RM, Buonerba F, Baroni M. Flavin monooxygenase metabolism: why medicinal chemists should matter. J Med Chem 2014; 57:6183-96. [PMID: 25003501 DOI: 10.1021/jm5007098] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
FMO enzymes (FMOs) play a key role in the processes of detoxification and/or bioactivation of specific pharmaceuticals and xenobiotics bearing nucleophilic centers. The N-oxide and S-oxide metabolites produced by FMOs are often active metabolites. The FMOs are more active than cytochromes in the brain and work in tandem with CYP3A4 in the liver. FMOs might reduce the risk of phospholipidosis of CAD-like drugs, although some FMOs metabolites seem to be neurotoxic and hepatotoxic. However, in silico methods for FMO metabolism prediction are not yet available. This paper reports, for the first time, a substrate-specificity and catalytic-activity model for FMO3, the most relevant isoform of the FMOs in humans. The application of this model to a series of compounds with unknown FMO metabolism is also reported. The model has also been very useful to design compounds with optimal clearance and in finding erroneous literature data, particularly cases in which substances have been reported to be FMO3 substrates when, in reality, the experimentally validated in silico model correctly predicts that they are not.
Collapse
Affiliation(s)
- Gabriele Cruciani
- Laboratory for Chemoinformatics and Molecular Modelling, Department of Chemistry, Biology and Biotechnology, University of Perugia , Via Elce di Sotto 8, 06123 Perugia, Italy
| | | | | | | | | | | |
Collapse
|
10
|
Rudraiah S, Rohrer PR, Gurevich I, Goedken MJ, Rasmussen T, Hines RN, Manautou JE. Tolerance to acetaminophen hepatotoxicity in the mouse model of autoprotection is associated with induction of flavin-containing monooxygenase-3 (FMO3) in hepatocytes. Toxicol Sci 2014; 141:263-77. [PMID: 24973094 DOI: 10.1093/toxsci/kfu124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acetaminophen (APAP) pretreatment with a hepatotoxic dose (400 mg/kg) in mice results in resistance to a second, higher dose (600 mg/kg) of APAP (APAP autoprotection). Recent microarray work by our group showed a drastic induction of liver flavin containing monooxygenase-3 (Fmo3) mRNA expression in our mouse model of APAP autoprotection. The role of liver Fmo3, which detoxifies xenobiotics, in APAP autoprotection is unknown. The purpose of this study was to characterize the gene regulation and protein expression of liver Fmo3 during APAP hepatotoxicity. The functional consequences of Fmo3 induction were also investigated. Plasma and livers were collected from male C57BL/6J mice over a period of 72 h following a single dose of APAP (400 mg/kg) to measure Fmo3 mRNA and protein expression. Although Fmo3 mRNA levels increased significantly following APAP treatment, protein expression changed marginally. In contrast, both Fmo3 mRNA and protein expression were significantly higher in APAP autoprotected livers. Unlike male C57BL/6J mice, female mice have ∼80-times higher constitutive Fmo3 mRNA levels and are highly resistant to APAP hepatotoxicity. Coadministration of APAP with the FMO inhibitor methimazole rendered female mice susceptible to APAP hepatotoxicity, with no changes in susceptibility detected in male mice. Furthermore, a human hepatocyte cell line (HC-04) clone over-expressing human FMO3 showed enhanced resistance to APAP cytotoxicity. Taken together, these findings establish for the first time induction of Fmo3 protein expression and function by xenobiotic treatment. Our results also indicate that Fmo3 expression and function plays a role in protecting the liver from APAP-induced toxicity. Although the mechanism(s) of this protection remains to be elucidated, this work describes a novel protective function for this enzyme.
Collapse
Affiliation(s)
- Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Philip R Rohrer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Igor Gurevich
- Cellular Dynamics International, Madison, Wisconsin 53711
| | - Michael J Goedken
- Rutgers University, Office of Translational Science, New Brunswick, New Jersey 08901
| | - Theodore Rasmussen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Ronald N Hines
- US EPA, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina 27711
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| |
Collapse
|
11
|
Veeravalli S, Omar BA, Houseman L, Hancock M, Gonzalez Malagon SG, Scott F, Janmohamed A, Phillips IR, Shephard EA. The phenotype of a flavin-containing monooyxgenase knockout mouse implicates the drug-metabolizing enzyme FMO1 as a novel regulator of energy balance. Biochem Pharmacol 2014; 90:88-95. [PMID: 24792439 DOI: 10.1016/j.bcp.2014.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/17/2014] [Accepted: 04/18/2014] [Indexed: 11/28/2022]
Abstract
Flavin-containing monooxygenases (FMOs) of mammals are thought to be involved exclusively in the metabolism of foreign chemicals. Here, we report the unexpected finding that mice lacking Fmos 1, 2 and 4 exhibit a lean phenotype and, despite similar food intake, weigh less and store less triglyceride in white adipose tissue (WAT) than wild-type mice. This is a consequence of enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure (REE). This is fuelled, in part, by increased fatty acid β-oxidation in skeletal muscle, which would contribute to depletion of lipid stores in WAT. The enhanced energy expenditure is attributed, in part, to an increased capacity for exercise. There is no evidence that the enhanced REE is due to increased adaptive thermogenesis; instead, our results are consistent with the operation in WAT of a futile energy cycle. In contrast to FMO2 and FMO4, FMO1 is highly expressed in metabolic tissues, including liver, kidney, WAT and BAT. This and other evidence implicates FMO1 as underlying the phenotype. The identification of a novel, previously unsuspected, role for FMO1 as a regulator of energy homeostasis establishes, for the first time, a role for a mammalian FMO in endogenous metabolism. Thus, FMO1 can no longer be considered to function exclusively as a xenobiotic-metabolizing enzyme. Consequently, chronic administration of drugs that are substrates for FMO1 would be expected to affect energy homeostasis, via competition for endogenous substrates, and, thus, have important implications for the general health of patients and their response to drug therapy.
Collapse
Affiliation(s)
- Sunil Veeravalli
- 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
| | - Matthew Hancock
- 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
| | - Azara Janmohamed
- Institute of Structural and Molecular Biology, 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
|
12
|
Frick A, Suzuki O, Butz N, Chan E, Wiltshire T. In vitro and in vivo mouse models for pharmacogenetic studies. Methods Mol Biol 2014; 1015:263-78. [PMID: 23824862 DOI: 10.1007/978-1-62703-435-7_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
The identification of causative genes underlying biomedically relevant phenotypes, particularly complex multigenic traits, is of vital interest to modern medicine. Using genome-wide association analysis, many studies have successfully identified thousands of loci (called quantitative trait loci or QTL), some of these associating with drug response phenotypes. However, the determination and validation of putative genes has been much more challenging. The actions of drugs, both efficacious and deleterious, are complex phenotypes that are controlled or influenced in part by genetic mechanisms.Investigation for genetic correlates of complex traits and pharmacogenetic traits is often difficult to perform in human studies due to cost, availability of relevant sample population, and limited ability to control for environmental effects. These challenges can be circumvented with the use of mouse models for pharmacogenetic studies. In addition, the mouse can be treated at sub- and supratherapeutic doses and subjected to invasive procedures, which can facilitate measures of drug response phenotypes, making identification of pharmacogenetically relevant genes more feasible. The availability of multiple mouse genetic and phenotypic resources is an additional benefit to using the mouse for pharmacogenetic studies.Here, we describe the contribution of animal models, specifically the mouse, towards the field of pharmacogenetics. In this chapter, we describe different mouse models, including the knockout mouse, recombinant mouse inbred strains, in vitro mouse cell-based assays, as well as novel experimental approaches like the Collaborative Cross recombinant mouse inbred panel, which can be applied to preclinical pharmacogenetics research. These approaches can be used to assess drug response phenotypes that are difficult to model in humans, thereby facilitating drug discovery, development, and application.
Collapse
Affiliation(s)
- Amber Frick
- Division of Pharmacotherapy and Experimental Therapeutics, Institute for Pharmacogenomics and Individualized Therapy, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | |
Collapse
|
13
|
Watson JD, Prokopec SD, Smith AB, Okey AB, Pohjanvirta R, Boutros PC. TCDD dysregulation of 13 AHR-target genes in rat liver. Toxicol Appl Pharmacol 2014; 274:445-54. [DOI: 10.1016/j.taap.2013.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 12/20/2022]
|
14
|
Abstract
The pharmacokinetics of the atypical antipsychotic, olanzapine, display large interindividual variation leading to multiple-fold differences in drug exposure between patients at a given dose. This variation in turn gives rise to the need for individualized dosing in order to avoid concentration-dependent adverse effects or therapeutic failure. Genetically determined differences in olanzapine metabolism represent a less studied source of variability in comparison to environmental and physiological factors. In this review, we summarize available in vitro and in vivo data addressing the influence of polymorphisms in drug-metabolizing enzymes on olanzapine serum exposure. The polymorphic CYP2D6 enzyme appears to have no significant influence on olanzapine steady-state serum concentrations. The formation of the various olanzapine metabolites is influenced by polymorphisms in the genes coding for CYP1A2, CYP1A expression regulator AHR, UGT1A4 and UGT2B10, as well as FMO3. An impact on steady-state olanzapine serum concentrations has been suggested for variants of CYP1A2 and UGT1A4, with somewhat conflicting findings. The potential involvement of FMO1 and CYP3A43 in olanzapine disposition has also been suggested but needs future validation.
Collapse
Affiliation(s)
- Mao Mao Söderberg
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | - Marja-Liisa Dahl
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden.
| |
Collapse
|
15
|
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]
|
16
|
Palmer AL, Leykam VL, Larkin A, Krueger SK, Phillips IR, Shephard EA, Williams DE. Metabolism and pharmacokinetics of the anti-tuberculosis drug ethionamide in a flavin-containing monooxygenase null mouse. Pharmaceuticals (Basel) 2012; 5:1147-59. [PMID: 23580869 PMCID: PMC3621790 DOI: 10.3390/ph5111147] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/08/2012] [Accepted: 10/16/2012] [Indexed: 11/16/2022] Open
Abstract
Multiple drug resistance (MDR) in Mycobacterium tuberculosis (mTB), the causative agent for tuberculosis (TB), has led to increased use of second-line drugs, including ethionamide (ETA). ETA is a prodrug bioactivated by mycobacterial and mammalian flavin-containing monooxygenases (FMOs). FMO2 is the major isoform in the lungs of most mammals, including primates. In humans a polymorphism exists in the expression of FMO2. FMO2.2 (truncated, inactive) protein is produced by the common allele, while the ancestral allele, encoding active FMO2.1, has been documented only in individuals of African and Hispanic origin, at an incidence of up to 50% and 7%, respectively. We hypothesized that FMO2 variability in TB-infected individuals would yield differences in concentrations and ratios of ETA prodrug and metabolites. In this study we assessed the impact of the FMO2 genetic polymorphism on the pharmacokinetics of ETA after administration of a single oral dose of ETA (125 mg/kg) to wild type and triple Fmo1/2/4-null mice, measuring levels of prodrug vs. metabolites in plasma collected from 0 to 3.5 h post-gavage. All mice metabolized ETA to ETA S-oxide (ETASO) and 2-ethyl-4-amidopyridine (ETAA). Wild type mice had higher plasma concentrations of metabolites than of parent compound (p = 0.001). In contrast, Fmo1/2/4-null mice had higher plasma concentrations of parent compound than of metabolites (p = 0.0001). Thus, the human FMO2 genotype could impact the therapeutic efficacy and/or toxicity of ETA.
Collapse
Affiliation(s)
- Amy L. Palmer
- Department of Environmental and Molecular Toxicology, Oregon State University, 1007 ALS Corvallis, OR 97331, USA; (A.L.P.); (V.L.L.); (A.L.)
| | - Virginia L. Leykam
- Department of Environmental and Molecular Toxicology, Oregon State University, 1007 ALS Corvallis, OR 97331, USA; (A.L.P.); (V.L.L.); (A.L.)
| | - Andrew Larkin
- Department of Environmental and Molecular Toxicology, Oregon State University, 1007 ALS Corvallis, OR 97331, USA; (A.L.P.); (V.L.L.); (A.L.)
| | - Sharon K. Krueger
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Institute Corvallis, OR 97331, USA;
| | - Ian R. Phillips
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK;
| | - Elizabeth A. Shephard
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK;
| | - David E. Williams
- Department of Environmental and Molecular Toxicology, Oregon State University, 1007 ALS Corvallis, OR 97331, USA; (A.L.P.); (V.L.L.); (A.L.)
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Institute Corvallis, OR 97331, USA;
| |
Collapse
|
17
|
Abstract
BACKGROUND Cigarette smoking and other forms of tobacco use are the leading cause of preventable mortality in the world. A better understanding of the etiology of nicotine addiction may help to increase the success rate of cessation and to decrease the massive morbidity and mortality associated with smoking. METHODS To identify genetic polymorphisms that contribute to nicotine dependence, our group undertook a genetic association study including three enzyme families that potentially influence nicotine metabolism: cytochrome P450 enzymes, flavin monooxygenases (FMOs), and UDP-glucuronosyl transferases. RESULTS Several polymorphisms in FMO1 showed association in a discovery sample, and were tested in an independent replication sample. One polymorphism, rs10912765, showed an association that remained significant after Bonferroni correction (nominal P=0.0067, corrected P=0.0134). Several additional polymorphisms in linkage disequilibrium with this single nucleotide polymorphism also showed association. Subsequent in-vitro experiments characterized FMO1 as a more efficient catalyst of nicotine N-oxidation than FMO3. In adult humans, FMO1 is primarily expressed in the kidney and is likely to be a major contributor to the renal metabolism and clearance of therapeutic drugs. FMO1 is also expressed in the brain and could contribute to the nicotine concentration in this tissue. CONCLUSION These findings suggest that polymorphisms in FMO1 are significant risk factors in the development of nicotine dependence and that the mechanism may involve variation in nicotine pharmacology.
Collapse
|
18
|
Shephard EA, Phillips IR. The potential of knockout mouse lines in defining the role of flavin-containing monooxygenases in drug metabolism. Expert Opin Drug Metab Toxicol 2010; 6:1083-94. [DOI: 10.1517/17425255.2010.503705] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Madrigal-Bujaidar E, Cárdenas García Y, Álvarez-González I. Chromosomal aberrations induced by imipramine and desipramine in mouse. Hum Exp Toxicol 2010; 29:297-302. [DOI: 10.1177/0960327110361751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Imipramine (IMI) and desipramine (DES) are two drugs widely used for the treatment of depression as well as for other diseases. In the present study, we determined their capacity to induce chromosomal aberrations in mouse bone marrow cells. Three doses of each compound were tested and their results were compared with the frequency of chromosomal aberrations obtained in a control group as well as with a group treated with cyclophosphamide. Our results showed a significant increase in chromosome damage with the doses tested for each compound: 7, 20, and 60 mg/kg in the case of IMI, and 2, 20, and 60 mg/kg as regards DES. This last drug induced stronger chromosomal damage than IMI. Our results agree with previous studies regarding the induction of micronuclei and sister chromatid exchanges by the drugs in mouse and suggest caution with respect to their use in long-term treatments.
Collapse
Affiliation(s)
| | - Y. Cárdenas García
- Laboratorio de Genética. Escuela Nacional de Ciencias Biologicas, IPN, Mexico, Escuela Superior de Medicina IPN, Mexico
| | - I. Álvarez-González
- Laboratorio de Genética. Escuela Nacional de Ciencias Biologicas, IPN, Mexico
| |
Collapse
|