Note
Potential for drug interactions mediated by polymorphic flavin-containing monooxygenase 3 in human livers

https://doi.org/10.1016/j.dmpk.2014.09.008Get rights and content

Abstract

Human flavin-containing monooxygenase 3 (FMO3) in the liver catalyzes a variety of oxygenations of nitrogen- and sulfur-containing medicines and xenobiotic substances. Because of growing interest in drug interactions mediated by polymorphic FMO3, benzydamine N-oxygenation by human FMO3 was investigated as a model reaction. Among the 41 compounds tested, trimethylamine, methimazole, itopride, and tozasertib (50 μM) suppressed benzydamine N-oxygenation at a substrate concentration of 50 μM by approximately 50% after co-incubation. Suppression of N-oxygenation of benzydamine, trimethylamine, itopride, and tozasertib and S-oxygenation of methimazole and sulindac sulfide after co-incubation with the other five of these six substrates was compared using FMO3 proteins recombinantly expressed in bacterial membranes. Apparent competitive inhibition by methimazole (0–50 μM) of sulindac sulfide S-oxygenation was observed with FMO3 proteins. Sulindac sulfide S-oxygenation activity of Arg205Cys variant FMO3 protein was likely to be suppressed more by methimazole than wild-type or Val257Met variant FMO3 protein was. These results suggest that genetic polymorphism in the human FMO3 gene may lead to changes of drug interactions for N- or S-oxygenations of xenobiotics and endogenous substances and that a probe battery system of benzydamine N-oxygenation and sulindac sulfide S-oxygenation activities is recommended to clarify the drug interactions mediated by FMO3.

Introduction

The flavin-containing monooxygenases (FMOs, EC 1.14.13.8) are a family of NADPH-dependent enzymes that catalyze the oxygenation of a wide variety of nucleophilic compounds containing a nitrogen, sulfur, phosphorous, or selenium atom [1], [2]; such compounds include the anti-inflammatory drug benzydamine, antithyroid drug methimazole, gastroprokinetic agent itopride, hereditary polyposis drug sulindac sulfide, anti-cancer agent tozasertib, and diet-derived trimethylamine (Fig. 1) [3], [4], [5]. Histamine H2-receptor antagonist ranitidine [6], dipeptidyl peptidase IV inhibitor teneligliptin [7], α7 neuronal nicotinic receptor agonist AZD0328 [8], and peroxisome proliferator-activated receptor dual agonist MK-0767 [9] are listed as medicinal and new drug candidate substrates of FMO.

FMO3 is considered the prominent functional FMO form expressed in adult human liver [10], [11]. Human protein-coding gene FMO3 and its mRNA expression levels are given in http://www.ncbi.nlm.nih.gov/UniGene/under [UniGene 240387 – Hs.445350]. Genetic polymorphism of FMO3 [12], [13], [14] and/or post-translational modification by environmental factors such as nitric oxide could cause interindividual differences in FMO3 levels or FMO3 catalytic function [15], [16]. Loss-of-function mutations, nonsense mutations, and missense mutations of FMO3 [17], [18], [19] produce phenotypes associated with the inherited disorder trimethylaminuria (also known as fish odor syndrome). In the literature, reported mutations in the FMO3 gene are given using systematic and trivial names [18]. In the course of identification of novel mutations of FMO3 and functional analysis in Japanese individuals suffering from trimethylaminuria, we found common and unique FMO3 variants [19]. We previously reported that Val257Met FMO3 protein had almost the same activity as wild-type FMO3, but [Glu158Lys; Glu308Gly] and Arg205Cys FMO3 proteins exhibited slightly and moderately decreased activities, respectively, with respect to N-oxygenation of trimethylamine [19].

FMOs form a complementary enzyme system to the cytochrome P450 enzymes and have been found to oxygenate several soft, highly polarizable nucleophilic heteroatom-containing chemicals and several drugs analyzed [20]. It has previously been suggested that the products of FMO3-mediated metabolism are generally benign, highly polar, and readily excreted [20]. The absence of induction of FMO3 by xenobiotics is strikingly different to the case for cytochrome P450 enzymes, which are induced by a number of xenobiotics [1]. FMO has been shown to exhibit a stable 4a-flavin hydroperoxide intermediate capable of oxygenating both nucleophiles and electrophiles in its catalytic cycle, even in the absence of oxygenatable substrate [21]. In this context, little information regarding apparent drug interactions has been reported so far for FMO3 [22]. Some of the preclinical research and development areas related to FMOs are not yet fully mature. Therefore, in the present study, we investigated the effects of a variety of N- and S-containing chemicals on benzydamine N-oxygenation to test its suitability as an index reaction for the enzymatic activity of recombinant human FMO3. Further investigations were carried out on FMO3-mediated oxidations of trimethylamine, methimazole, itopride, and tozasertib (selected as strong suppressors of benzydamine N-oxygenation) and sulindac sulfide (selected as typical S-containing clinical substrate). Strong suppression by S-containing methimazole on sulindac sulfide S-oxygenation by polymorphic FMO3 was observed. The present results suggest that a battery system of benzydamine N-oxygenation and sulindac sulfide S-oxygenation activities would be useful as probe substrate reactions to clarify drug interactions mediated via polymorphic human FMO3.

Section snippets

Chemicals and enzymes

Benzydamine hydrochloride, itopride, methimazole, sulindac sulfoxide, and sulindac sulfide were purchased from Sigma–Aldrich (St. Louis, MO, USA) and trimethylamine from Wako Pure Chemicals (Osaka, Japan). Tozasertib was purchased from Cayman Chemical (Ann Arbor, MI, USA). Benzydamine N-oxide was a generous gift from Prof. Allan E. Rettie (University of Washington). Recombinant human FMO3 proteins were prepared as reported previously [23]. In the preliminary trials, the use of human livers for

Results and discussion

Because of growing interest in drug interactions of candidate medicines [3], [4], [5], [6], [7], [8], [9] mediated by polymorphic FMO3, we investigated the suppression of FMO3-mediated benzydamine N-oxygenation by N- and S-containing chemicals (Fig. 2). Benzydamine (50 μM) was selected as a marker reaction for wild-type human FMO3 in the presence of 50 μM (open bars) or 100 μM (closed bars) of a total of 41 effector chemicals. Trimethylamine, methimazole, itopride, and tozasertib (50 μM)

Acknowledgments

The authors thank John Cashman, Tomomi Taniguchi-Takizawa, Norie Murayama, and Kuniharu Sasaki for their technical assistance and David Smallbones for his English language advice. This work was supported in part by the Ministry of Education, Culture, Sports, Science and Technology of Japan (25460198).

References (29)

  • J.R. Cashman et al.

    Human flavin-containing monooxygenases

    Annu Rev Pharmacol Toxicol

    (2006)
  • J.R. Cashman et al.

    Population-specific polymorphisms of the human FMO3 gene: significance for detoxication

    Drug Metab Dispos

    (2000)
  • D.M. Ziegler

    An overview of the mechanism, substrate specificities, and structure of FMOs

    Drug Metab Rev

    (2002)
  • C.S. Park et al.

    Ethnic differences in allelic frequency of two flavin-containing monooxygenase 3 (FMO3) polymorphisms: linkage and effects on in vivo and in vitro FMO activities

    Pharmacogenetics

    (2002)
  • Cited by (20)

    • Different substrate elimination rates of model drugs pH-dependently mediated by flavin-containing monooxygenases and cytochromes P450 in human liver microsomes

      2021, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      After incubation for the designated periods at five time points, reactions were terminated by transferring a 100-μL aliquot of reaction mixture to a stop solution (methanol or acetonitrile) that contained internal standards. The concentrations of these substrates in the resulting supernatants centrifuged at 10,000 g for 5 min at 4 °C were determined using liquid chromatography–tandem mass spectrometry [6,8,9]. The clearance values were calculated from the slope of the remaining substrate versus time curve determined by linear regression analysis.

    • In vivo drug interactions of itopride and trimethylamine mediated by flavin-containing monooxygenase 3 in humanized-liver mice

      2021, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      However, little information is available regarding apparent drug interactions mediated by FMO3. Some preclinical research and development activities are aimed at investigating FMO-dependent drug interactions in vitro [11]; however, these procedures have not yet been fully implemented. Itopride is in wide clinical use and is also valuable as a typical FMO3-dependent probe drug substrate [12,13].

    • Ligand stabilization and effect on unfolding by polymorphism in human flavin-containing monooxygenase 3

      2020, International Journal of Biological Macromolecules
      Citation Excerpt :

      Human flavin-containing monooxygenases (hFMOs) comprise a family of five isoenzymes and are the second most important phase 1 drug-metabolizing enzymes after cytochromes P450 [1–3]. Its isoform 3 (hFMO3) is predominantly expressed in the liver where substrates containing nitrogen-, sulphur- and phosphorous-containing soft nucleophiles [4–12] are transformed into more polar and excretable metabolites [13–18]. Wild type hFMO3 contributes to the metabolism of several important drugs [19] such as ranitidine, cimetidine, tamoxifen, clozapine, benzydamine.

    • Monoamine Oxidases and Flavin-Containing Monooxygenases

      2018, Comprehensive Toxicology: Third Edition
    • Identification of structural determinants of NAD(P)H selectivity and lysine binding in lysine N<sup>6</sup>-monooxygenase

      2016, Archives of Biochemistry and Biophysics
      Citation Excerpt :

      These enzymes catalyze hydroxylation, sulfoxidation, epoxidation, Baeyer–Villiger oxidation, and halogenation reactions through the incorporation of a single oxygen atom into a variety of substrates [34,35]. They have received great attention in environmental, industrial, and pharmaceutical applications due to their regio- and enantioselective catalytic activity and their wide-spectrum of substrate reactivity [36–38]. NMOs are members of the class B flavin monooxygenases and catalyze the hydroxylation of the side chain amine of l-Lys or l-Orn.

    View all citing articles on Scopus
    View full text