Mammalian flavin-containing monooxygenases: structure/function, genetic polymorphisms and role in drug metabolism

https://doi.org/10.1016/j.pharmthera.2005.01.001Get rights and content

Abstract

Flavin-containing monooxygenase (FMO) oxygenates drugs and xenobiotics containing a “soft-nucleophile”, usually nitrogen or sulfur. FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. FMO and CYP also exhibit similar tissue and cellular location, molecular weight, substrate specificity, and exist as multiple enzymes under developmental control. The human FMO functional gene family is much smaller (5 families each with a single member) than CYP. FMO does not require a reductase to transfer electrons from NADPH and the catalytic cycle of the 2 monooxygenases is strikingly different. Another distinction is the lack of induction of FMOs by xenobiotics.

In general, CYP is the major contributor to oxidative xenobiotic metabolism. However, FMO activity may be of significance in a number of cases and should not be overlooked. FMO and CYP have overlapping substrate specificities, but often yield distinct metabolites with potentially significant toxicological/pharmacological consequences.

The physiological function(s) of FMO are poorly understood. Three of the 5 expressed human FMO genes, FMO1, FMO2 and FMO3, exhibit genetic polymorphisms. The most studied of these is FMO3 (adult human liver) in which mutant alleles contribute to the disease known as trimethylaminuria. The consequences of these FMO genetic polymorphisms in drug metabolism and human health are areas of research requiring further exploration.

Introduction

Mammalian flavin-containing monooxygenase (FMO, EC 1.14.13.8) was first described by Dr. Daniel Ziegler and colleagues at the University of Texas at Austin (reviewed in Ziegler, 1980, Ziegler, 1988, Ziegler, 1991, Ziegler, 1993, Ziegler, 2002, Poulsen & Ziegler, 1995). This hepatic microsomal enzyme system was demonstrated to utilize oxygen and NADPH to convert N,N′-dimethylaniline to the N-oxide (Ziegler & Pettit, 1964, Mitchell & Zieger, 1969). The location, co-factor requirement and activity of FMO toward xenobiotics were very similar to the then recently characterized cytochrome P450 (CYP) monooxygenase and for a time this enzyme was known as “Ziegler's enzyme”. In 1971, the enzyme was purified from porcine liver microsomes and definitively demonstrated to be a distinct monooxygenase containing flavin, with no heme (Ziegler et al., 1971a, Ziegler & Mitchell, 1972, Ziegler & Poulsen, 1978). Again, based on studies on the catalytic activity of this enzyme, it was referred to as “mixed-function amine oxidase”, a term that did not capture its wide substrate range.

FMO is a flavoprotein containing a single FAD. Much of the early work describing the structure/function, mechanism, regulation, and substrate specificity of FMO was with the purified porcine liver enzyme, again led by Dr. Dan Ziegler and his colleague Dr. Larry Poulsen (Ziegler et al., 1971a, Ziegler et al., 1971b, Ziegler et al., 1992, Poulsen & Ziegler, 1979, Poulsen & Ziegler, 1995). The pig liver enzyme oxygenates a wide range of sulfur- and nitrogen-containing xenobiotics and, in some cases, also oxygenates selenium, iodine, boron, phosphorus and even carbon. An unusual feature of this class of enzymes is that substrate binding has no effect on velocity. In general, any chemical containing a soft nucleophile that gains access to the peroxyflavin intermediate (see Section 2.1 for details) is a potential substrate.

In 1984, the first direct evidence of the existence of multiple forms of FMO was obtained when the enzyme was purified from rabbit lung microsomes independently by 2 laboratories (Williams et al., 1984, Tynes et al., 1985). The “lung” FMO was distinct from the “liver” FMO in having high activity toward primary alkyl amines, restricted substrate specificity related to steric properties, resistance to detergent inhibition and enhanced thermal stability (Williams et al., 1984, Williams et al., 1985, Williams et al., 1990, Tynes & Hodgson, 1985a, Tynes & Hodgson, 1985b, Tynes et al., 1985, Tynes et al., 1986, Poulsen et al., 1986, Nagata et al., 1990, Hodgson & Levi, 1991, Venkatesh et al., 1992). Since those early studies, FMOs have been purified and/or cloned from multiple sources (Table 1). Until recently, it was thought that only 5 genes were expressed in mammals (Hines et al., 1994, Lawton et al., 1994, Cashman, 1995, Phillips et al., 1995). A sixth human gene, FMO6, identified by the Sanger Center chromosome 1 sequencing project, was demonstrated to be a psuedogene (Hines et al., 2002). These FMO genes are located in a cluster on the long arm of chromosome 1 (q23–25). The laboratories of Drs. Ian Phillips and Elizabeth Shephard have recently discovered a second FMO gene cluster, located also on chromosome 1. This second cluster in humans contains an additional 5 genes, all of which appear to be pseudogenes. Interestingly, in the mouse, this second gene cluster contains 3 genes which do not appear to be pseudo genes (Hernandez et al., 2004).

As with the CYP monooxygenase system, over the course of evolutionary plant–animal warfare, FMO developed broad substrate specificity at the expense of turnover. Both monooxygenases are capable of oxidizing thousands of plant alkaloids and other natural products as well as the thousands of synthetic therapeutic drugs. The CYP turnover number for most xenobiotics is 1–20 min−1 and the number is slightly higher for FMO, i.e., 30–60 min−1. Although active toward many of the same substrates, CYP and FMO often produce distinct metabolites. It is recognized in the drug development field that potential concerns can arise if a drug contains a structural feature capable of being bioactivated by CYP to a toxic metabolite, e.g., an epoxide, oxon, or primary aryl N-hydroxy-amine. In general, FMO oxygenation results in metabolites with reduced pharmacological and toxicological properties. As with every rule, there are exceptions. FMO can S-oxygenate thioureas to sulfenic acids which can undergo redox cycling or be further converted to the reactive sulfinic acid (Poulsen et al., 1979, Neal & Halpert, 1982, Ziegler, 1982, Krieter et al., 1984, Sabourin & Hodgson, 1984, Nagata et al., 1990, Decker & Doerge, 1991, Guo & Ziegler, 1991, Guo et al., 1992, Onderwater et al., 1999, Onderwater et al., 2004, Kim & Ziegler, 2000, Smith & Crespi, 2002, Henderson et al., 2004b).

The structure/function properties of the FMO family are still relatively unknown, as are the physiological function(s) of this enzyme (other than in xenobiotic metabolism), the impact of polymorphic expression, and the importance, relative to CYP, in the metabolism and efficacy of therapeutic drugs. In this review, we have attempted to summarize the current state of that knowledge and suggest possible avenues of further research.

Section snippets

Catalytic mechanism and reactive oxygen species ROS generation

FMO belongs to a class of monooxygenases capable of generating a stable C4a peroxyflavin intermediate (Massey, 1994). This intermediate is stable for minutes to hours at 4 °C and can be observed spectrally (Beaty & Ballou, 1980, Ziegler et al., 1980, Williams et al., 1984, Jones & Ballou, 1986). In the first step of the catalytic cycle, FAD undergoes 2-electron reduction by NADPH (Fig. 1). The reduced flavin reacts rapidly with molecular oxygen to form the peroxyflavin and it is in this state,

Genetic variants in flavin-containing monooxygenase 3 and trimethylaminuria

Trimethylaminuria (TMAU), previously referred to as “Fish-Odor Syndrome”, is manifested clinically by a body odor resembling rotten fish and is accompanied by various psychosocial abnormalities (Ayesh & Smith, 1990, Treacy et al., 1998, Mitchell & Smith, 2001). This disease is classified by NIH as a rare disease and has been the focus of 2 international meetings sponsored by the NIH Office of Rare Diseases. The actual incidence of TMAU, however, may be underreported. Due to the paucity of

Role of flavin-containing monooxygenase in drug metabolism

FMO and CYP share a number of similarities with respect to tissue, cellular, and organelle expression, the utilization of oxygen and NADPH as cofactors, and activity toward many of the same drugs. In order to determine in vitro the relative contribution of FMO versus CYP in the oxygenation of a particular drug, a number of approaches have been used. The activity of any and all microsomal CYPs can be inhibited by titration with antibody to NADPH CYP reductase, carbon monoxide, or addition of

Conclusions and suggestions for future research

The flavin-containing monooxygenase (FMO) is a family of enzymes that like the cytochrome P450 (CYP) superfamily evolved during plant–animal warfare to metabolize a plethora of xenobiotics. There are numerous similarities between these monooxygenase systems. Both utilize reducing equivalents from NADPH to reduce the second atom of molecular oxygen to water and incorporate the other atom into the substrate. FMO and CYP are between 50 and 60 kDa, located in the endoplasmic reticulum and are

References (314)

  • J.R. Cashman et al.

    Microsomal oxidation of thiobenzamide. A photometric assay for the flavin-containing monooxygenase

    Biochem Biophys Res Commun

    (1981)
  • J.R. Cashman et al.

    Role of hepatic flavin-containing monooxygenase in drug and chemical metabolism in adult humans

    Chem Biol Interact

    (1995)
  • M.J. Cheesman et al.

    Critical role of histidine residues in cyclohexanone monooxygenase expression, cofactor binding and catalysis

    Chem Biol Interact

    (2003)
  • G.-P. Chen et al.

    Liver microsome and flavin-containing monooxygenase catalyzed oxidation of organic selenium compounds

    Arch Biochem Biophys

    (1994)
  • K. Chiba et al.

    N-oxygenation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine by the rat liver flavin-containing monooxygenase expressed in yeast cells

    Eur J Pharmacol

    (1995)
  • E. Chieli et al.

    Role of the microsomal FAD-containing monooxygenase in the liver toxicity of thioacetamide S-oxide

    Toxicology

    (1984)
  • W.G. Chung et al.

    Oxidation of caffeine to theobromine and theophylline is catalyzed primarily by flavin-containing monooxygenase in liver microsomes

    Biochem Biophys Res Commun

    (1997)
  • C.J. Decker et al.

    Covalent binding of 14C- and 35S-labelled thiocarbamides in rat hepatic microsomes

    Biochem Pharmacol

    (1992)
  • T.R. Devereux et al.

    N-oxidation and demethylation of N,N-dimethylaniline by rabbit liver and lung microsomes: effects of age and metals

    Chem Biol Interact

    (1974)
  • C.T. Dolphin et al.

    The flavin-containing monooxygenase 2 gene (FMO2) of humans, but not of other primates, encodes a truncated, nonfunctional protein

    J Biol Chem

    (1998)
  • R.J. Duescher et al.

    Flavin-containing monooxygenase (FMO)-dependent metabolism of methionine and evidence for FMO3 being the major FMO involved in methionine sulfoxidation in rabbit liver and kidney microsomes

    J Biol Chem

    (1994)
  • A.A. Elfarra

    Potential role of the flavin-containing monooxygenases in the metabolism of endogenous compounds

    Chem Biol Interact

    (1995)
  • H. Engler et al.

    Mechanism of inactivation of thyroid peroxidase by thioureylene drugs

    Biochem Pharmacol

    (1982)
  • J.G. Falls et al.

    Molecular cloning, sequencing, and expression in Escherichia coli of mouse flavin-containing monooxygenase 3 (FMO3): comparison with the human isoform

    Arch Biochem Biophys

    (1997)
  • M.B. Fisher et al.

    Prochiral sulfide probes for the active-site topography of rabbit flavin-containing monooxygenase

    Tetrahedron: Asymmetry

    (1997)
  • M.W. Fraaije et al.

    Identification of a Baeyer–Villiger monooxygenase sequence motif

    FEBS Lett

    (2002)
  • D.E. Goeger et al.

    Oxidation of dimethylselenide to dimethylselenoxide by microsomes from rat liver and lung and by flavin-containing monooxygenase from pig liver

    Arch Biochem Biophys

    (1994)
  • S.J. Grossman et al.

    Thiobenzamide-induced hepatotoxicity: effects of substituents and route of administration on the nature and extent of liver injury

    Toxicol Appl Pharmacol

    (1991)
  • W.X. Guo et al.

    Use of thiocarbamides as selective substrate probes for isoforms of flavin-containing monooxygenases

    Biochem Pharmacol

    (1992)
  • I. Gut et al.

    Trimethylamine N-oxygenation and N-demethylation in rat liver microsomes

    Biochem Pharmacol

    (1993)
  • N.P. Hajjar et al.

    Sulfoxidation of thioether-containing pesticides by the flavin–adenine dinucleotide-dependent monooxygenase of pig liver microsomes

    Biochem Pharmacol

    (1982)
  • M.A. Hamman et al.

    Stereoselective sulfoxidation of sulindac sulfide by flavin-containing monooxygenases. Comparison of human liver and kidney microsomes and mammalian enzymes

    Biochem Pharmacol

    (2000)
  • R.P. Hanzlik et al.

    Relative hepatoxtoxicity of thiobenzamides and thiobenzamides-S-oxides in the rat

    Toxicol Appl Pharmacol

    (1980)
  • S.J. Hardwick et al.

    Protection of rats against the effects of alpha-naphthylthiourea (ANTU) by elevation of non-protein sulphydryl levels

    Biochem Pharmacol

    (1991)
  • M.C. Henderson et al.

    S-oxygenation of the thioether organophosphate insecticides phorate and disulfoton by human lung flavin-containing monooxygenase 2

    Biochem Pharmacol

    (2004)
  • B.R. Akerman et al.

    Two novel mutations of the FMO3 gene in a proband with trimethylaminuria

    Hum Mutat

    (1999)
  • M. Attar et al.

    Cytochrome P450 2C8 and flavin-containing monooxygenases are involved in the metabolism of tazarotenic acid in humans

    Drug Metab Dispos

    (2003)
  • T.K. Attwood et al.

    PRINTS and its automatic supplement, prePRINTS

    Nucleic Acids Res

    (2003)
  • M. Barber et al.

    Abnormalities of flavin monooxygenase as an etiology for sideroblastic anemia

    Am J Hematol

    (2000)
  • T. Basarab et al.

    Sequence variations in the flavin-containing monooxygenase 3 gene (FMO3) in fish odour syndrome

    Br J Dermatol

    (1999)
  • E.B. Belldina et al.

    Steady-state pharmacokinetics and pharmacodynamics of cysteamine bitartrate in paediatric nephropathic cystinosis patients

    Br J Clin Pharmacol

    (2003)
  • S. Bhamre et al.

    Cerebral flavin-containing monooxygenase-mediated metabolism of antidepressants in brain: immunochemical properties and immunocytochemical localization

    J Pharmacol Exp Ther

    (1993)
  • J.E. Biaglow et al.

    Factors influencing the oxidation of cysteamine and other thiols: implications for hyperthermic sensitization and radiation protection

    Radiat Res

    (1984)
  • B.L. Blake et al.

    Metabolism of thioridazine by microsomal monooxygenases: relative roles of P450 and flavin-containing monooxygenase

    Xenobiotica

    (1995)
  • M.G. Boersma et al.

    Role of cytochromes P-450 and flavin-containing monooxygenase in the biotransformation of 4-fluoro-N-methylaniline

    Drug Metab Dispos

    (1993)
  • M.R. Boyd et al.

    Studies on the mechanism of toxicity and of development of tolerance to the pulmonary toxin, alpha-naphthylthiourea (ANTU)

    Drug Metab Dispos

    (1976)
  • E.B. Brittebo

    Metabolism-dependent toxicity of methimazole in the olfactory nasal mucosa

    Pharmacol Toxicol

    (1995)
  • A. Brunelle et al.

    Characterization of two human flavin-containing monooxygenase (form 3) enzymes expressed in Escherichia coli as maltose binding protein fusions

    Drug Metab Dispos

    (1997)
  • T. Buronfosse et al.

    Stereoselective sulfoxidation of the pesticide methiocarb by flavin-containing monooxygenase and cytochrome P450-dependent monooxygenases of rat liver microsomes. Anticholinesterase activity of the two sulfoxide enantiomers

    J Biochem Toxicol

    (1995)
  • E.J. Butler et al.

    Trimethylamine and fishy taint in eggs

    World Poult Sci J

    (1984)
  • Cited by (475)

    • The desert woodrat (Neotoma lepida) induces a diversity of biotransformation genes in response to creosote bush resin

      2024, Comparative Biochemistry and Physiology Part - C: Toxicology and Pharmacology
    • Molecular and functional characterization of flavin-containing monooxygenases (FMO1–6) in tree shrews

      2024, Comparative Biochemistry and Physiology Part - C: Toxicology and Pharmacology
    View all citing articles on Scopus
    View full text