Elsevier

Biochemical Pharmacology

Volume 49, Issue 5, 1 March 1995, Pages 591-602
Biochemical Pharmacology

Research paper
Particular ability of cytochromes P450 3A to form inhibitory P450-iron-metabolite complexes upon metabolic oxidation of aminodrugs

https://doi.org/10.1016/0006-2952(94)00477-4Get rights and content

Abstract

The ability of 21 drugs containing an amine function to form inhibitory P450-iron-metabolite complexes absorbing around 455 nm was studied on liver microsomes from rats treated with various P450 inducers. These drugs belong to different chemical and therapeutic series and exhibit very different structures. In the case of eight compounds (diltiazem, lidocaine, imipramine, SKF 525A, fluoxetine, l-α-acetylmethadol, methadol and desmethyltamoxifen) whose oxidation by microsomes from rats treated with several inducers was studied, only dexamethasone (DEX)-treated rat microsomes and, to a lesser extent, phenobarbital (PB)-treated rat microsomes, were able to give significant amounts of 455 nm absorbing complexes. Ten of the 21 compounds studied gave such complexes with DEX-treated rat microsomes, while only three compounds gave complexes (in low amounts) with PB-treated rat microsomes only. For all compounds leading to complexes both with DEX- and PB-treated rat microsomes, much higher amounts of complexes were obtained with DEX-treated rat microsomes. DEX-treated rat microsomes also led to the most intense type I spectral interactions with most of the compounds studied, and very often exhibited the highest N-dealkylation activities towards the tertiary or secondary amine function of the drugs used. A few exceptions aside, there generally exists a qualitative relationship between the ability of P450 3As, induced by DEX, to bind and N-dealkylate amino compounds and their propensity to lead to 455 nm absorbing complexes. This was confirmed by in vivo experiments showing that rats treated with diltiazem, tamoxifen or imipramine accumulated large amounts of 455 nm absorbing complexes in their liver only after pretreatment with DEX and, to a lesser extent, with PB. This particular ability of P450 3As to oxidize amino drugs with formation of inhibitory P450-metabolite complexes could be of great importance for the appearance of drug interactions in man.

References (53)

  • JP Renaud et al.

    Particular ability of liver P450s3A to catalyze the oxidation of N-(omega)-hydroxyarginine to citrulline and nitrogen oxides and occurrence in NO synthases of a sequence very similar to the hemebinding sequence in P450s

    Biochem Biophys Res Commun

    (1993)
  • MR Franklin

    Inhibition of mixed function oxidations by substrates forming reduced cytochrome P-450 metabolic-intermediate complexes

    Pharmacol Ther

    (1977)
  • B Lindeke et al.

    N-dealkylation and deamination

  • JB Schenkman et al.

    Diethylaminoethyl, 2,2-diphenylvalerate HC1 (SKF 525A). In vivo and in vitro effects of metabolism by rat liver microsomes, formation of an oxygenated complex

    Biochem Pharmacol

    (1972)
  • M Delaforge et al.

    Dual effects of macrolide antibiotics on rat liver cytochrome P-450. Induction and formation of metabolite complexes: a structure-activity relationship

    Biochem Pharmacol

    (1983)
  • M Murray

    Complexation of cytochrome P450 isozymes in hepatic microsomes from SKF 525 A-induced rats

    Arch Biochem Biophys

    (1988)
  • D Larrey et al.

    Formation of an inactive cytochrome P-450 Fe(II)-metabolite complex after administration of amiodarone in rats, mice and hamsters

    Biochem Pharmacol

    (1986)
  • M Murray et al.

    Inhibition and metabolite complexation of rat hepatic microsomal cytochrome P-450 by tricyclic antidepressants

    Biochem Pharmacol

    (1992)
  • OH Lowry et al.

    Protein measurement with the Folin phenol reagent

    J Biol Chem

    (1951)
  • M Delaforge et al.

    The cytochrome P-450 metabolite complex derived from troleandomycin: properties in vitro and stability in vivo

    Chem-Biol Interact

    (1984)
  • DR Nelson et al.

    The P450 superfamily—update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature

    DNA Cell Biol

    (1993)
  • FP Guengerich et al.

    Comparison of levels of several human microsomal cytochrome P-450 enzymes and epoxide hydrolase in normal and disease states using immunochemical analysis of surgical liver samples

    J Pharmacol Exp Ther

    (1991)
  • J De Waziers et al.

    Cytochrome P-450 isoenzymes, epoxide hydrolase and glutathione transferases in rat and human hepatic and extrahepatic tissues

    J Pharmacol Exp Ther

    (1990)
  • GI Murray et al.

    The immunocytochemical localization and distribution of cytochrome P-450 in normal human hepatic and extrahepatic tissues with a monoclonal antibody to human cytochrome P-450

    Br J Clin Pharmacol

    (1988)
  • DM Heuman et al.

    Immunochemical evidence for induction of a common form of hepatic cytochrome P-450 in rats treated with pregnenolone-16 α-carbonitrile or other steroidal or non steroidal agents

    Mol Pharmacol

    (1982)
  • SA Wrighton et al.

    Identification of the cytochrome P450 induced by macrolide antibiotics in rat liver as the glucocorticoid responsive cytochrome P450

    Biochemistry

    (1985)
  • Cited by (104)

    • The mechanisms of interactions of psychotropic drugs with liver and brain cytochrome P450 and their significance for drug effect and drug-drug interactions

      2022, Biochemical Pharmacology
      Citation Excerpt :

      On the other hand, some psychotropics, such as tricyclic antidepressants, phenothiazine neuroleptics or haloperidol produce reactive metabolites, which irreversibly bind to CYP enzymes, in this way inhibiting enzyme activity (Fig. 3). Tricyclic antidepressants and fluoxetine have been shown to form inhibitory CYP-iron(II)-nitrosoalkane metabolite complexes, affecting CYP3A, CYP2C11 and CYP2A [57,58]. Likewise, epoxide intermediates of imipramine (and probably of other antidepressants) formed during hydroxylation process, radical cations of phenothiazine neuroleptics mediating their sulfoxidation and ring hydroxylation, and haloperidol pyridinium metabolites (HPP+) irreversibly inhibit CYP2D6 activity [51,59,60].

    • The nitrosoamphetamine metabolite is accommodated in the active site of human hemoglobin: Spectroscopy and crystal structure

      2020, Journal of Inorganic Biochemistry
      Citation Excerpt :

      A hydrogen bond was evident between the nitroso O-atom of the AmphNO and the distal His63 residue (Fig. 6B), with the nitroso NO bond nearly eclipsing an Fe–N(heme) bond (with a N(por)–Fe–N–O torsion angle of ~2°). As we note in the Introduction, nitrosoalkane binding to the Fe centers of heme proteins are known to inhibit the functions of these proteins [8,13–24]. To verify the Fe oxidation states extant in the crystal obtained from the Hb/AmphNHOH reaction, we retrieved the crystal used in the X-ray diffraction data collection and obtained its UV–vis spectrum (after dissolving the used crystal in buffer).

    • Time-dependent enzyme inactivation: Numerical analyses of in vitro data and prediction of drug-drug interactions

      2020, Pharmacology and Therapeutics
      Citation Excerpt :

      The amine group is hydroxylated and further oxidized to form a nitroso group that coordinates with the heme. The heme is then reduced to a more stable ferrous form (Bensoussan et al., 1995; Mansuy, Gans, Chottard, & Bartoli, 1977) with an absorbance maximum of 448 to 456 nm (Franklin, 1977). The MIC is stable and can be isolated (Ekroos & Sjögren, 2006; Murray & Reidy, 1990).

    • Effect of antidepressant drugs on cytochrome P450 2C11 (CYP2C11) in rat liver

      2013, Pharmacological Reports
      Citation Excerpt :

      Like in rats, the investigated antidepressants are not regarded as potent inhibitors of human CYP2C9 [19, 33, 43]. As has been mentioned elsewhere, tricyclic antidepressants and fluoxetine can form reactive/intermediate metabolites, which irreversibly inactivate a few CYP isoforms (CYP2C11, CYP2D, CYP3A and CYP2A) after prolonged incubation in vitro with high concentrations of antidepressants, or after in vivo administration of high doses of the drugs [5, 27, 28, 30, 31], as well as when used at therapeutic concentrations in vivo after a 24-h exposure to antidepressants [11, 12, 18]. The present results suggest that CYP2C11 inactivation via this mechanism by imipramine, desipramine and fluoxetine (used in therapeutic concentrations) via this mechanism is also possible, since a one-day treatment with pharmacological doses of the above-mentioned antidepressants diminishes (down to 66–70% of the control values) the enzyme activity in rats (Model II).

    • Heterocycle-substituted proline dipeptides as potent VLA-4 antagonists

      2010, Bioorganic and Medicinal Chemistry Letters
    View all citing articles on Scopus
    View full text