Elsevier

Biochemical Pharmacology

Volume 80, Issue 6, 15 September 2010, Pages 903-912
Biochemical Pharmacology

The relative importance of CYP26A1 in hepatic clearance of all-trans retinoic acid

https://doi.org/10.1016/j.bcp.2010.05.023Get rights and content

Abstract

All-trans retinoic acid (RA) is a critical signaling molecule and its concentration is tightly regulated. Several P450 enzymes including CYP26A1, CYP2C8, and CYP3A4 have been proposed to be responsible for RA clearance in the liver but their quantitative importance has not been demonstrated. To determine the contribution of CYP26A1 to hepatic clearance of RA, CYP26A1 protein was quantified in 37 human liver microsomes (HLMs). CYP26A1 expression ranged from not detectable to 2.80 pmol/mg microsomal protein. RA clearance by P450 enzymes abundant in human liver was measured in Supersomes®. CYP2C8, CYP3A4, CYP3A5 and CYP3A7 metabolized RA with unbound Km values of 3.4–7.2 μM and Vmax values of 2.3–4.9 pmol/min/pmol P450, but were less efficient than CYP26A1 in clearing RA. Simulations performed for livers with varying P450 expression levels over a range of RA concentrations demonstrated that at both endogenous and therapeutic concentrations of RA, CYP26A1 is the primary enzyme responsible for 4-OH RA formation clearance. HLM incubation data showed that 4-OH RA formation velocity varied from 0.2 to 15.3 pmol/min/mg microsomal protein and velocity in HLMs was significantly correlated (p < 0.01) to CYP26A1, CYP3A4, and CYP3A5 protein content, but not to CYP2C8. When experimental data were scaled to in vivo clearances, the predicted hepatic clearance of RA (0.07 L/min using combined Supersome® data) was similar to the published in vivo clearance of RA. These findings suggest that CYP26A1 is the P450 isoform that should be targeted when designing RA metabolism blocking agents.

Introduction

All-trans retinoic acid (RA) is the biologically active metabolite of Vitamin A (Retinol). RA serves as a critical signaling molecule, regulating gene transcription, cell division and apoptosis during development and adult life [1], [2], [3]. Both too much and too little RA have been linked with detrimental effects, including increased risk of infection leading to death [4], decreased fertility [5], Alzheimer's disease [6] and developmental defects [7]. Therefore, concentrations of RA must be tightly regulated.

Cellular exposure to RA is regulated by controlled synthesis and metabolism. The liver plays a role in maintaining RA homeostasis by contributing to RA synthesis and clearance, serving as the major storage organ for retinyl esters and RA, and by providing a first pass barrier for dietary intake of RA [8]. The liver is also likely to be the main organ clearing therapeutically administered RA. The exact metabolic pathway by which RA is cleared from the body is still unclear, but the primary oxidative pathway for RA clearance is believed to be via P450 mediated formation of 4-OH RA [9], [10], [11]. There is an abundance of data showing the oxidation of RA to 4-OH RA and other metabolites, including a subsequent oxidation to 4-oxo RA, in in vitro systems such as COS-1, V79-4, and HeLa cells; liver, lung, kidney, and testis microsomes and recombinant P450 expression systems [9], [10], [11], [12], [13], [14], [15], [16]. In all of these studies, 4-OH RA was detected as the primary oxidation product formed from RA. Despite the in vitro data, the identity of in vivo metabolites of RA is still mainly qualitative and the only metabolite detected in plasma is the 4-oxo RA. 4-oxo RA has been detected in rabbits administered RA or retinyl palmitate [17] and in human plasma [18]. At present the identity of the enzyme forming 4-oxo RA is unknown. However, since 4-OH RA is a necessary primary metabolite leading to 4-oxo RA, the presence of 4-oxo RA in vivo demonstrates that 4-OH RA is also formed in vivo.

Several P450s have been demonstrated to oxidize RA in vitro, including CYP2C8, CYP2C9, CYP2C18, CYP3A4, CYP3A5, CYP3A7, and the newer family of P450s, CYP26. [9], [10], [11], [19] The general consensus of the previously published studies evaluating the importance of P450 isoforms in the metabolism of RA is that CYP2C8 and members of the CYP3A subfamily are the primary P450s responsible for RA clearance. However, the proposed relative importance of these enzymes varies and published data suggests that other enzymes may be involved in RA metabolism as well. Quinidine, a classic selective inhibitor of CYP2D6, caused a 50% reduction in 4-OH RA formation in human liver microsomes despite the fact that recombinant CYP2D6 did not form 4-OH RA from RA [11]. In another study, ketoconazole (20 μM) inhibited 50% of the 4-hydroxylation activity indicating that CYP3A4 contribution to RA hydroxylation does not exceed 50% [10]. These findings suggest that enzymes other than CYP3A4 and CYP2C8 may be important for RA hydroxylation in human liver. A possible additional enzyme would be a member of the CYP26 family, as the contribution of CYP26 enzymes to hepatic RA oxidation was not analyzed in these previous studies [20].

CYP26A1 was first identified in 1996 and has been demonstrated to be inducible by RA and to metabolize RA [13]. The enzymes of the CYP26 family, including CYP26A1 as well as CYP26B1 [21] and CYP26C1 [22], are believed to be the primary CYP enzymes responsible for clearing RA based on the fact that these enzymes metabolize RA efficiently in vitro and are inducible by RA [23]. Cyp26a1−/− and Cyp26b1−/− mice are not viable [24], [25] and show developmental defects similar to the teratogenic effects associated with excess RA [24]. Studies examining mRNA expression have identified CYP26A1 mRNA in the human liver [26], [27] whereas CYP26B1 and CYP26C1 mRNA were present in only a fraction of livers and at a lower level of expression than CYP26A1. Based on mRNA data CYP26A1 was suggested to be the major liver CYP26 isoform [27].

The goal of this study was to determine the role of CYP26A1 in hepatic clearance of RA. We hypothesized that if CYP26A1 is expressed in the liver, it will be the major contributor to RA clearance in this organ, due to the efficiency with which CYP26A1 metabolizes RA [19]. We also hypothesized that due to the low Km of RA towards CYP26A1, with increasing exposure to RA during therapeutic treatment with RA, CYP26A1 will be saturated and the role of other P450 isoforms in RA clearance will increase. To test this hypothesis, RA hydroxylation kinetics was determined for individual P450 isoforms and the relative importance of these enzymes to RA clearance was predicted. Using human liver microsomes (HLMs) from multiple donors, the expression of CYP26A1 protein in the liver was quantified, and RA hydroxylation measured in multiple donors. The obtained data was used in simulating the contribution of CYP26A1 to RA clearance for the purpose of establishing the relative importance of CYP26A1 between individuals and over a variety of RA concentrations.

Section snippets

Reagents

RA, acitretin, cholate, imidazole and NADPH were purchased from Sigma–Aldrich (St. Louis, MO). 4-OH RA was synthesized in-house as previously described [19]. Supersomes® were purchased from Gentest (BD Biosciences, Woburn, MA.) All supersomes were co-expressed with reductase and, with the exception of CYP4A11, CYP1A1, CYP1A2, CYP2C18 and CYP2D6, with cytochrome b5. Purified CYP1A1 protein was purchased from Panvera (Madison, WI). The remaining purified proteins were obtained as gifts from the

Quantification of CYP26 in human liver microsomes

To determine the relative importance of CYP26A1 in RA clearance, the amount of CYP26A1 in a panel of livers was quantified. The specificity and sensitivity of the commercial and in-house antibodies was validated by evaluating them against two CYP26 insect cell membrane preparations and a panel of supersomes of ten P450s abundant in human liver. Cross reactivity with specific P450 isoforms was further confirmed using seven purified CYP proteins. Supplemental Figure 1 shows the ability of the

Discussion

The enzymes in the CYP26 family have been suggested to be the main RA hydroxylases in mammals and other chordates. Based on previously published RNA work, CYP26A1 appears to be the main liver CYP26 isoform [26]. CYP26B1 mRNA was not quantifiable in 46% of livers in the donor bank used in this study, and when detected, the amounts were significantly lower than CYP26A1 mRNA [27]. CYP26A1 has been previously demonstrated to be very efficient in metabolizing RA and has a high affinity to RA [19].

Acknowledgments

This work was supported in part by NIH grants T32 GM007750, P01 GM32165, and R01 GM081569. While the cytochrome P450 concentrations for CYP2C8, CYP3A4, and CYP3A5 used in the predictions for individual HLMs have been previously reported [32], [33], we thank Dr. Rheem Totah, University of Washington, Department of Medicinal Chemistry and Dr. Yvonne Lin, University of Washington, Department of Pharmaceutics for providing us a matched list of the concentrations to the specific HLM donor number so

References (46)

  • G. Tzimas et al.

    The role of metabolism and toxicokinetics in retinoid teratogenesis

    Curr Pharm Des

    (2001)
  • J. Marill et al.

    Retinoic acid metabolism and mechanism of action: a review

    Curr Drug Metab

    (2003)
  • G. Duester

    Retinoic acid synthesis and signaling during early organogenesis

    Cell

    (2008)
  • World_Health_Organization. Macronutrient deficiencies: vitamin A deficiency. [cited 2009 April 4]; Available from:...
  • S.B. Wolbach et al.

    Tissue changes following deprivation of fat-soluble A vitamin

    Exp Med

    (1925)
  • K. Shudo et al.

    Towards retinoid therapy for Alzheimer's disease

    Curr Alzheimer Res

    (2009)
  • R. Blomhoff et al.

    Overview of retinoid metabolism and function

    J Neurobiol

    (2006)
  • J.L. Napoli

    Biochemical pathways of retinoid transport, metabolism, and signal transduction

    Clin Immunol Immunopathol

    (1996)
  • J. Marill et al.

    Identification of human cytochrome P450s involved in the formation of all-trans-retinoic acid principal metabolites

    Mol Pharmacol

    (2000)
  • L.C. McSorley et al.

    Identification of human cytochrome P450 isoforms that contribute to all-trans-retinoic acid 4-hydroxylation

    Biochem Pharmacol

    (2000)
  • L. Nadin et al.

    Participation of CYP2C8 in retinoic acid 4-hydroxylation in human hepatic microsomes

    Biochem Pharmacol

    (1999)
  • C.A. Frolik et al.

    Isolation and identification of 4-hydroxy- and 4-oxoretinoic acid. In vitro metabolites of all-trans-retinoic acid in hamster trachea and liver

    Biochemistry

    (1979)
  • J.A. White et al.

    Identification of the retinoic acid-inducible all-trans-retinoic acid 4-hydroxylase

    J Biol Chem

    (1996)
  • H. Chen et al.

    Catalysis of the 4-hydroxylation of retinoic acids by cyp3a7 in human fetal hepatic tissues

    Drug Metab Dispos

    (2000)
  • J.V. Chithalen et al.

    HPLC-MS/MS analysis of the products generated from all-trans-retinoic acid using recombinant human CYP26A

    J Lipid Res

    (2002)
  • P.D. Fiorella et al.

    Microsomal retinoic acid metabolism. Effects of cellular retinoic acid-binding protein (type I) and C18-hydroxylation as an initial step

    J Biol Chem

    (1994)
  • G. Tzimas et al.

    Embryotoxic doses of vitamin A to rabbits result in low plasma but high embryonic concentrations of all-trans-retinoic acid: risk of vitamin A exposure in humans

    J Nutr

    (1996)
  • T.E. Gundersen et al.

    Quantitative high-throughput determination of endogenous retinoids in human plasma using triple-stage liquid chromatography/tandem mass spectrometry

    Rapid Commun Mass Spectrom

    (2007)
  • J.D. Lutz et al.

    Expression and functional characterization of cytochrome P450 26A1, a retinoic acid hydroxylase

    Biochem Pharmacol

    (2009)
  • J.E. Thatcher et al.

    The role of CYP26 enzymes in retinoic acid clearance

    Expert Opin Drug Metab Toxicol

    (2009)
  • J.A. White et al.

    Identification of the human cytochrome P450. P450RAI-2, which is predominantly expressed in the adult cerebellum and is responsible for all-trans-retinoic acid metabolism

    Proc Nat Acad Sci USA

    (2000)
  • M. Taimi et al.

    A novel human cytochrome P450, CYP26C1, involved in metabolism of 9-cis and all-trans isomers of retinoic acid

    J Biol Chem

    (2004)
  • S.J. Lee et al.

    The discovery of new coding alleles of human CYP26A1 that are potentially defective in the metabolism of all-trans retinoic acid and their assessment in a recombinant cDNA expression system

    Pharmacogenet Genomics

    (2007)
  • Cited by (64)

    • Application of reaction phenotyping to address pharmacokinetic variability in patient populations

      2023, Overcoming Obstacles in Drug Discovery and Development: Surmounting the Insurmountable-Case Studies for Critical Thinking
    • New luciferin-based probe substrates for human CYP26A1

      2020, Biochemistry and Biophysics Reports
      Citation Excerpt :

      Interestingly, strong expression of CYP26A1 is correlated with poorer survival in breast cancer patients [3]. In the human adult liver, CYP26A1 is clearly the most strongly expressed CYP26 enzyme and also the predominant atRA hydroxylase [4]. Therefore, selective inhibitors of CYP26A1 are of pharmacologic interest as they could decrease the systemic clearance of retinoic acid (see below).

    View all citing articles on Scopus
    View full text