Review
The marmoset cytochrome P450 superfamily: Sequence/phylogenetic analyses, genomic structure, and catalytic function

https://doi.org/10.1016/j.bcp.2019.113721Get rights and content

Abstract

The common marmoset (Callithrix jacchus) is a New World monkey that has attracted much attention as a potentially useful primate model for preclinical testing. A total of 36 marmoset cytochrome P450 (P450) isoforms in the P450 1–51 subfamilies have been identified and characterized by the application of genome analysis and molecular functional characterization. In this mini-review, we provide an overview of the genomic structures, sequence identities, and substrate selectivities of marmoset P450s compared with those of human P450s. Based on the sequence identity, phylogeny, and genomic organization of marmoset P450s, orthologous relationships were established between human and marmoset P450s. Twenty-four members of the marmoset P450 1A, 2A, 2B, 2C, 2D, 2E, 3A, 4A, and 4F subfamilies shared high degrees of homology in terms of cDNA (>89%) and amino acid sequences (>85%) with the corresponding human P450s; P450 2C76 was among the exceptions. Phylogenetic analysis using amino acid sequences revealed that marmoset P450s in the P450 1–51 families were located in the same clades as their human and macaque P450 homologs. This finding underlines the evolutionary closeness of marmoset P450s to their human and macaque homologs. Most marmoset P450 1–4 enzymes catalyzed the typical drug-metabolizing reactions of the corresponding human P450 homologs, except for some differences of P450 2A6 and 2B6. Consequently, it appears that the substrate specificities of enzymes in the P450 1–4 families are generally similar in marmosets and humans. The information presented here supports a better understanding of the functional characteristics of marmoset P450s and their similarities and differences with human P450s. It is hoped that this mini-review will facilitate the successful use of marmosets as primate models in drug metabolism and pharmacokinetic studies.

Introduction

Because of their close genetic and physiological similarities to humans, nonhuman primates are important species for evaluating the safety and therapeutic efficacy of candidates for drug development. The common marmoset (Callithrix jacchus), a New World monkey, is a potentially useful primate in efficacy/pharmacology, safety/toxicology, and drug metabolism and pharmacokinetic studies for preclinical testing. The common marmoset has various advantages, such as genetic closeness to humans, small size, fast maturation, high fertility, and ease of handling [1]. Marmosets have been favored for biomedical research in fields such as neurobiology [2], immunology [3], obesity [4], and aging [5]. The cynomolgus macaque (Macaca fascicularis), an Old World monkey, has been widely used as a preclinical model in drug metabolism and pharmacokinetic studies because of its availability and its long history of use as a laboratory animal species.

Cytochromes P450 (P450) constitute a superfamily of heme-thiolate enzymes that mediate the biotransformation and metabolism of endogenous steroid hormones, xenobiotics, and drugs. Currently known human P450 isoforms are encoded by 57 functional genes and 58 pseudogenes [6]. P450 enzymes in the P450 1A, 2B, 2C, 2D, 2E, and 3A subfamilies are major drug-metabolizing enzymes [7], whereas those in the P450 4 family are involved in the metabolism of endogenous [8] and exogenous compounds [9]. P450 enzymes can catalyze various types of enzymatic reactions, including hydroxylation, dealkylation, sulfoxidation, epoxidation, and N-oxidation [10]. Interspecies differences of P450-mediated drug metabolism have been identified in commonly used preclinical animal species, including mice, rats, dogs, pigs, and cynomolgus macaques [11]. Consequently, to select appropriate animal species for drug metabolism and pharmacokinetic studies, it is important to understand the orthologous relationships and functional characteristics of their P450 isoforms.

In 2013, the Brain Mapping by Integrated Neurotechnologies for Disease Studies (Brain/MINDS) project was set up. Its aims were to accelerate the understanding of human brain disorders using marmosets and to facilitate the use of marmosets for drug development [12]. However, despite the importance of the marmoset as a preclinical nonhuman primate species, only limited numbers of P450 isoforms have been identified, including P450 1A2, 2B6, 2C8, 2D6, 2E1, 3A4, 3A5, and 3A90. We participated in the Brain/MINDS project to investigate the characteristics of drug metabolism and pharmacokinetics in marmosets. By using transcriptome data of marmoset tissues obtained by next-generation high-throughput RNA sequencing analysis [13] and by using marmoset genome data based on whole-genome shotgun sequencing and assembly [14], we identified and characterized a large number of marmoset P450 isoforms in the P450 1–51 families (see http://drnelson.uthsc.edu/CytochromeP450.html). Our understanding of P450-mediated drug metabolism has been accelerated by the comprehensive cDNA cloning and functional analyses of marmoset P450 isoforms carried out as part of the Brain/MINDS project. This mini-review compares the characteristics of the P450 1A, 2A, 2B, 2C, 2D, 2E, and 3A subfamilies in marmosets and humans.

Section snippets

Gene identification, cDNA cloning, and phylogenetic analysis of marmoset P450s

Marmoset P450 151 gene clusters were analyzed based on marmoset genome data in the UCSC Genome Browser (https://genome.ucsc.edu/index.html) (Fig. 1). The locations and directions of most marmoset P450 151 genes in gene clusters are well conserved with those of human P450 genes. However, the gene arrangements of the P450 2ABFGST, 2C, 2D, 3A, 4ABXZ, and 4F clusters are slightly different between humans and marmosets. In the P450 1 family, gene arrangements in the P450 1A, 1B, and 1D clusters

Conclusions and future perspectives

This mini-review provides a comprehensive description of the gene organization, sequence identity, phylogeny, tissue distribution, induction, and substrate specificity of marmoset P450 1–4 family members and makes comparisons with human P450 isoforms. Several remarkable characteristics of marmoset P450 1–4 family members are revealed, including the partial differences of genomic organization in the P450 2ABFGST, 2C, 2D, 3A, 4ABXZ, and 4F clusters; the similarities of P450 1A1/1A2 and 3A4

CRediT authorship contribution statement

Shotaro Uehara: Formal analysis, Investigation. Yasuhiro Uno: Investigation. Hiroshi Yamazaki: Writing - original draft, Writing - review & editing.

Acknowledgments

We thank Drs. Erika Sasaki, Takashi Inoue, Norie Murayama, and Makiko Shimizu for their support with the experiments. The authors also greatly thank David Smallbones for copyediting a draft of this article. This work resulted from the “Construction of System for Spread of Primate Model Animals” initiative under the Strategic Research Program for Brain Sciences of the Japan Agency for Medical Research and Development.

Declaration of interest

The authors report no declarations of interest.

References (100)

  • S. Koskela et al.

    Expression of CYP2A genes in human liver and extrahepatic tissues

    Biochem. Pharmacol.

    (1999)
  • T.G. Schulz et al.

    Assessment of P450 induction in the marmoset monkey using targeted anti-peptide antibodies

    Biochim. Biophys. Acta

    (2001)
  • H. Raunio et al.

    Immunochemical and catalytical studies on hepatic coumarin 7-hydroxylase in man, rat, and mouse

    Biochem. Pharmacol.

    (1988)
  • K. Mayumi et al.

    Characterization of marmoset CYP2B6: cDNA cloning, protein expression and enzymatic functions

    Biochem. Pharmacol.

    (2013)
  • S. Narimatsu et al.

    Cloning of a cDNA encoding a novel marmoset CYP2C enzyme, expression in yeast cells and characterization of its enzymatic functions

    Biochem. Pharmacol.

    (2006)
  • K. Nakanishi et al.

    In vivo and in vitro diclofenac 5-hydroxylation mediated primarily by cytochrome P450 3A enzymes in common marmoset livers genotyped for P450 2C19 variants

    Biochem. Pharmacol.

    (2018)
  • S. Uehara et al.

    Oxidation of R- and S-omeprazole stereoselectively mediated by liver microsomal cytochrome P450 2C19 enzymes from cynomolgus monkeys and common marmosets

    Biochem. Pharmacol.

    (2016)
  • S. Narimatsu et al.

    Regio- and stereoselective oxidation of propranolol enantiomers by human CYP2D6, cynomolgus monkey CYP2D17 and marmoset CYP2D19

    Chem. Biol. Interact.

    (2011)
  • M.J. Paine et al.

    Residues glutamate 216 and aspartate 301 are key determinants of substrate specificity and product regioselectivity in cytochrome P450 2D6

    J. Biol. Chem.

    (2003)
  • P. Rowland et al.

    Crystal structure of human cytochrome P450 2D6

    J. Biol. Chem.

    (2006)
  • T.G. Schulz et al.

    Identification of CYP2E1 in marmoset monkey

    Biochim. Biophys. Acta

    (1998)
  • Y. Uno et al.

    Characterization of cynomolgus monkey cytochrome P450 (CYP) cDNAs: is CYP2C76 the only monkey-specific CYP gene responsible for species differences in drug metabolism?

    Arch. Biochem. Biophys.

    (2007)
  • S. Wu et al.

    Molecular cloning and expression of CYP2J2, a human cytochrome P450 arachidonic acid epoxygenase highly expressed in heart

    J. Biol. Chem.

    (1996)
  • J.M. Lasker et al.

    Formation of 20-hydroxyeicosatetraenoic acid, a vasoactive and natriuretic eicosanoid, in human kidney. Role of Cyp4F2 and Cyp4A11

    J. Biol. Chem.

    (2000)
  • P.K. Powell et al.

    Identification of CYP4A11 as the major lauric acid omega-hydroxylase in human liver microsomes

    Arch. Biochem. Biophys.

    (1996)
  • U. Hoch et al.

    Covalently linked heme in cytochrome p4504a fatty acid hydroxylases

    J. Biol. Chem.

    (2001)
  • E.A. Dierks et al.

    The catalytic site of cytochrome P4504A11 (CYP4A11) and its L131F mutant

    J. Biol. Chem.

    (1998)
  • T. Hashizume et al.

    cDNA cloning and expression of a novel cytochrome p450 (cyp4f12) from human small intestine

    Biochem. Biophys. Res. Commun.

    (2001)
  • P. Christmas et al.

    Alternative splicing determines the function of CYP4F3 by switching substrate specificity

    J. Biol. Chem.

    (2001)
  • X. Cui et al.

    A novel human cytochrome P450 4F isoform (CYP4F11): cDNA cloning, expression, and genomic structural characterization

    Genomics

    (2000)
  • H. Yamazaki et al.

    Roles of NADPH-P450 reductase and apo- and holo-cytochrome b5 on xenobiotic oxidations catalyzed by 12 recombinant human cytochrome P450s expressed in membranes of Escherichia coli

    Protein Expr. Purif.

    (2002)
  • H. Yamazaki et al.

    Progesterone and testosterone hydroxylation by cytochromes P450 2C19, 2C9, and 3A4 in human liver microsomes

    Arch. Biochem. Biophys.

    (1997)
  • V. Hirani et al.

    Expression of CYP4F2 in human liver and kidney: assessment using targeted peptide antibodies

    Arch. Biochem. Biophys.

    (2008)
  • R. Jin et al.

    Role of human CYP4F2 in hepatic catabolism of the proinflammatory agent leukotriene B4

    Arch. Biochem. Biophys.

    (1998)
  • Z. Tang et al.

    Human cytochrome P450 4F11: heterologous expression in bacteria, purification, and characterization of catalytic function

    Arch. Biochem. Biophys.

    (2010)
  • I. Messaoudi et al.

    Nonhuman primate models of human immunology

    Antioxid Redox Signal

    (2011)
  • S.D. Tardif et al.

    Body mass growth in common marmosets: toward a model of pediatric obesity

    Am. J. Phys. Anthropol.

    (2013)
  • S.D. Tardif et al.

    The marmoset as a model of aging and age-related diseases

    ILAR J.

    (2011)
  • D.R. Nelson et al.

    Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants

    Pharmacogenetics

    (2004)
  • J.A. Williams et al.

    Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios

    Drug Metab. Dispos.

    (2004)
  • K.Z. Edson et al.

    CYP4 enzymes as potential drug targets: focus on enzyme multiplicity, inducers and inhibitors, and therapeutic modulation of 20-hydroxyeicosatetraenoic acid (20-HETE) synthase and fatty acid omega-hydroxylase activities

    Curr. Top. Med. Chem.

    (2013)
  • T. Hashizume et al.

    Involvement of CYP2J2 and CYP4F12 in the metabolism of ebastine in human intestinal microsomes

    J. Pharmacol. Exp. Ther.

    (2002)
  • P.B. Danielson

    The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans

    Curr. Drug Metab.

    (2002)
  • M. Martignoni et al.

    Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction

    Expert Opin. Drug Metab. Toxicol.

    (2006)
  • D. Cyranoski

    Marmosets are stars of Japan's ambitious brain project

    Nature

    (2014)
  • M. Shimizu et al.

    Qualitative de novo analysis of full length cDNA and quantitative analysis of gene expression for common marmoset (Callithrix jacchus) transcriptomes using parallel long-read technology and short-read sequencing

    PLoS One

    (2014)
  • K. Sato et al.

    Resequencing of the common marmoset genome improves genome assemblies and gene-coding sequence analysis

    Sci. Rep.

    (2015)
  • S. Hu et al.

    Evolution of the CYP2ABFGST gene cluster in rat, and a fine-scale comparison among rodent and primate species

    Genetica

    (2008)
  • Y. Uno et al.

    CYP2G2, pseudogenized in human, is expressed in nasal mucosa of cynomolgus monkey and encodes a functional drug-metabolizing enzyme

    Drug Metab. Dispos.

    (2011)
  • Y. Yasukochi et al.

    Evolution of the CYP2D gene cluster in humans and four non-human primates

    Genes Genet. Syst.

    (2011)
  • Cited by (12)

    • Polymorphic cytochromes P450 in non-human primates

      2022, Advances in Pharmacology
      Citation Excerpt :

      CYP2D genes are arranged in the human genome differently than in the macaque and marmoset genomes, and CYP2D8, a pseudogene in humans, encodes a functional enzyme in macaques and marmosets (Uehara, Uno, & Yamazaki, 2020; Uno, Iwasaki, et al., 2011). For CYP3A genes, the number and arrangements of genes in the genome are well-conserved between humans and macaques, but not between humans and marmosets (Uehara, Uno, & Yamazaki, 2020; Uno, Iwasaki, et al., 2011). Marmoset CYP3A5 and CYP3A90, which are highly homologous to human CYP3A5, are functional enzymes, whereas the marmoset genome does not contain a CYP3A43 ortholog.

    • Molecular cloning and tissue distribution of marmoset thiopurine S-methyltransferase

      2020, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      Because of their genetic closeness to humans and their small body size, early sexual maturity, and ease of handling, marmosets are useful in the nonclinical stages of development of pharmaceutical products. Moreover, our recent reports have indicated that the catalytic functions and tissue expression patterns of major drug-metabolizing enzymes, such as cytochromes P450, are similar in marmosets and humans [2]. Thiopurine S-methyltransferase (TPMT) is a cytoplasmic transmethylase that catalyzes the methylation of sulfur atoms in anticancer and immunosuppressive thiopurine drugs, including azathioprine, 6-mercaptopurine, and thioguanine [3].

    • Expression of functional sulfotransferases (SULT) 1A1, 1A3, 1B1, 1C2, 1E1, and 2A1 in common marmosets

      2020, Biochemical Pharmacology
      Citation Excerpt :

      The common marmoset (Callithrix jacchus) is a nonhuman primate species that is often used in drug metabolism and pharmacokinetic studies. In marmosets, cytochromes P450, flavin-containing monooxygenases, UDP-glucuronosyltransferases, and glutathione S-transferases have been investigated to elucidate the differences and similarities between marmosets and humans [6–9]. Moreover, marmoset aldehyde oxidase [10], catechol-O-methyltransferase [11], monoamine oxidases A and B [12], and thiopurine S-methyltransferase [13], have also been described.

    View all citing articles on Scopus
    1

    These authors contributed equally.

    View full text