Elsevier

Biochemical Pharmacology

Volume 39, Issue 6, 15 March 1990, Pages 1067-1075
Biochemical Pharmacology

Reactivity of diflunisal acyl glucuronide in human and rat plasma and albumin solutions

https://doi.org/10.1016/0006-2952(90)90286-TGet rights and content

Abstract

Diflunisal acyl glucuronide (DAG) is a major metabolite of diflunisal (DF) in rats and humans. We have investigated the reactivity of DAG, in purified albumin solutions and plasma from both rat and human sources, along three interrelated pathways: rearrangement via acyl migration to yield positional isomers of DAG, hydrolysis of DAG and/or its isomers to liberate DF, and formation of covalent adducts of DF (via DAG and/or its isomers) with plasma protein. Two initial concentrations of DAG (ca. 50 and 10 μg DF equivalents/mL) were used throughout. In all incubations, the order of quantitative importance of the reactions was: rearrangement > hydrolysis >covalent binding. At pH 7.4 and 37°, degradation of DAG in albumin solutions (e.g. half-life ca. 95 min in fatty acid-free human serum albumin) was retarded in comparison to that found in buffer alone (half-life ca. 35 min). Degradation in unbuffered rat and human plasma containing heparin was comparable to that found in buffer. Maximal covalent binding to protein was achieved after 4–8 hr incubation, and was greatest for fatty acid-free human serum albumin (165ng DF/mg albumin). Thereafter, slow degradation of the adducts was observed. Formation of DF-plasma protein adducts in vivo was also found in rats and humans dosed with DF.

References (40)

  • F. Compernolle et al.

    Glucuronic acid conjugates of bilirubin-IXα in normal bile compared with post-obstructive bile. Transformation of the 1-O-acylglucuronides into 2-, 3-, and 4-O-acylglucuronides

    Biochem J

    (1978)
  • EM Faed et al.

    Separation of two conjugates of clofibric acid (CPIB) found in the urine of subjects taking clofibrate

    Clin Exp Pharmacol Physiol

    (1978)
  • HPA Illing et al.

    pH dependent formation of β-glucuronidase resistant conjugates from the biosynthetic ester glucuronide of isoxepac

    Biochem Pharmacol

    (1981)
  • J Hasegawa et al.

    Apparent intramolecular acyl migration of zomepirac glucuronide

    Drug Metab Dispos

    (1982)
  • FW Janssen et al.

    Metabolic formation of N- and O-glucuronides of 3-(p-chlorophenyl)thiazolo[3,2-a]benzimidazole — 2 — acetic acid. Rearrangement of the 1-O-acylglucuronide

    Drug Metab Dispos

    (1982)
  • RG Dickinson et al.

    pH-Dependent rearrangement of the biosynthetic ester glucuronide of valproic acid to β-glucuronidase-resistant forms

    Drug Metab Dispos

    (1984)
  • A Rachmel et al.

    Furosemide 1-O-acyl glucuronide. In vitro biosyn-thesis and pH-dependent isomerization to β-glucuronidase-resistant forms

    Drug Metab Dispos

    (1985)
  • J Caldwell et al.

    Acylation of amino acids and other endobiotics by xenobiotic carboxylic acids

  • ML Hyneck et al.

    Effect of pH on acyl migration and hydrolysis of tolmetin glucuronide

    Drug Metab Dispos

    (1988)
  • EM Faed

    Properties of acyi glucuronides: implications for studies of the pharmacokinetics and metabolism of acidic drugs

    Drug Metab Rev

    (1984)
  • Cited by (50)

    • Toxicological potential of acyl glucuronides and its assessment

      2017, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      In addition to the transacylation of 1-O-β-AG, ring-opened AGs (aldose), generated from positional isomers, also form covalent protein adducts, known as Schiff bases, followed by an Amadori rearrangement to form glycated proteins [52]. 1-O-β-AGs and their positional isomers of various carboxylic acid-containing drugs, such as zomepirac, diflunisal, etodolac, ketoprofen, and fibrates, covalently bind to proteins in human plasma and/or human serum albumin (HSA) [51,53–56]. The degradation rate constants of nine kinds of AGs in an incubation mixture containing HSA were strongly correlated with the levels of covalent binding to HSA (r2 = 0.995) [57].

    • Mechanistic role of acyl glucuronides

      2013, Drug-Induced Liver Disease
    • Chapter 3 Glucuronidation-Dependent Toxicity and Bioactivation

      2008, Advances in Molecular Toxicology
      Citation Excerpt :

      The ability of acyl glucuronides to form covalently bound adducts with endogenous macromolecules is now well documented and has been the subject of several excellent reviews [47,49,50]. The direct involvement of acyl glucuronides in the covalent modification of endogenous macromolecules has been demonstrated: (i) in vitro, by direct exposure of plasma proteins [59,77–81] or tissue microsomal protein [61,82] to acyl glucuronides, or by incubation of microsomal protein with carboxylic acid drugs in the presence and absence of glucuronidation co-factors [83,84]; (ii) in situ, using cultured hepatocytes exposed to carboxylic acid drugs in the presence and absence of glucuronidation inhibitors [85–87], or using control and UGT-expressing cell lines incubated with carboxylic acid drug [88,89] and (iii) in vivo, in rats administered carboxylic acid drugs with or without pre-treatment with glucuronidation inhibitors [90,91]. Elucidation of the chemical mechanisms of adduct formation has come primarily from in vitro studies utilising: (i) radiolabelled acyl glucuronide, labelled either on the drug or glucuronic acid moiety [59,92]; (ii) imine trapping reagents to investigate covalent binding via the glycation pathway [83,93–95] or (iii) more recently, with mass spectrometric analysis of covalently modified proteins and peptides [30–35,96–99].

    View all citing articles on Scopus
    View full text