Associate editor: R.A. Prough
Cytochrome P450 4F subfamily: At the crossroads of eicosanoid and drug metabolism

https://doi.org/10.1016/j.pharmthera.2006.03.008Get rights and content

Abstract

The cytochrome P450 4F (CYP4F) subfamily has over the last few years come to be recognized for its dual role in modulating the concentrations of eicosanoids during inflammation as well as in the metabolism of clinically significant drugs. The first CYP4F was identified because it catalyzed the hydroxylation of leukotriene B4 (LTB4) and since then many additional members of this subfamily have been documented for their distinct catalytic roles and functional significance. Recent evidence emerging in relation to the temporal change of CYP4F expression in response to injury and infection supports an important function for these isozymes in curtailing inflammation. Their tissue-dependent expression, isoform-based catalytic competence and unique response to the external stimuli imply a critical role for them to regulate organ-specific functions. From this standpoint variations in relative CYP4F levels in humans may have direct influence on the metabolic outcome through their ability to generate and/or degrade bioactive eicosanoids or therapeutic agents. This review covers the enzymatic characteristics and regulatory properties of human and rodent CYP4F isoforms and their physiological relevance to major pathways in eicosanoid and drug metabolism.

Introduction

Cytochrome P450s (CYP), phase I drug metabolizing enzymes, comprise a superfamily of heme-thiolate proteins that play critical roles in endogenous as well as xenobiotic metabolism (Guengerich, 1991, Oritz de Montellano, 1995, Anzenbacher and Anzenbacherova, 2001, Nebert and Russell, 2002). CYP-dependent monooxygenases are found predominantly in the microsomal fraction of tissues, but such activities can also be seen in the mitochondria (Anandatheerthavarada et al., 1997, Anandatheerthavarada et al., 1999, Robin et al., 2002). The microsomal CYP monooxygenase enzyme system, which is our interest here, has 3 major components: (a) the heme protein, CYP, (b) the flavoprotein, NADPH CYP-reductase and (c) phospholipids (Strobel et al., 1970, Coon, 2002, Coon, 2005). The reductase transfers 2 electrons to the CYP supplying reducing equivalents to the heme iron for reduction of molecular oxygen and product formation during catalysis (Strobel et al., 1995a).

Although first identified in liver, the presence and activity of CYP have been extended to extrahepatic tissues including kidney, colon, lung and brain (Guengerich and Mason, 1979, Strobel et al., 1997, Strobel et al., 2001, Ding and Kaminsky, 2003, Zhao and Imig, 2003, Kroetz and Xu, 2004). Currently, > 700 CYP have been characterized, inclusive of the many different species of organisms that have been studied (Nelson, 1999). These proteins are conveniently arranged into families and subfamilies on the basis of percent amino acid sequence identity (Nebert & Gonzalez, 1987). The individual CYP are represented by an Arabic number denoting the family, followed by an alphabetical letter designating the sub-family and an Arabic numeral representing the individual gene within the subfamily (Nelson et al., 1993, Nelson, 1999). This nomenclature is followed throughout this review.

Apart from having multiple forms, a fascinating fact about CYP is their broad substrate spectrum. While some CYP isoforms are fairly specific in their choice of substrate, such as those involved in steroidogenesis, the rest of them catalyze a large number of reactions.

Beyond their toxicological and pharmacological relevance, CYP enzymes play a central role in metabolizing endogenous products such as cholesterol, steroids, vitamins, eicosanoids and fatty acids (Capdevila et al., 1982, Andersson et al., 1989, McGiff, 1991, Kikuta et al., 1993, Kawashima et al., 1997, Lund et al., 1999, Node et al., 1999, Bylund et al., 2000, Capdevila et al., 2000, Thompson et al., 2000, Oliw et al., 2001, Nebert and Russell, 2002, Sontag and Parker, 2002, Cheng et al., 2004). Of particular interest is their role in the arachidonic acid (AA) cascade. AA, a polyunsaturated long-chain fatty acid esterified to cellular phospholipids, can be metabolized into various active and inactive products based on their cellular and physiological context. Prostaglandins (PG) and leukotrienes (LT) are the most widely studied metabolites of AA; however, several CYP in the subfamilies 2 and 4 catalyze conversion of AA into 20-hydroxyeicosatetraenoic acid (HETE) and epoxyeicosatrienoic acids (EET) (Fisslthaler et al., 1999, Capdevila et al., 2000, Lasker et al., 2000, Zeldin, 2001, Chuang et al., 2004, Kroetz and Xu, 2004). The focus of this review is the CYP4F subfamily which plays a pivotal role in metabolism of AA, its derivatives and many clinically important drugs.

Section snippets

History and discovery of cytochrome P450 4F (CYP4F) subfamily

CYP4F belong to a relatively newer CYP subfamily and were first discovered as part of an effort to define an inactivation pathway for leukotriene B4 (LTB4), a powerful mediator of inflammation. In the early 1980s, Hansson et al. (1981) were the first to report that in a 2-step breakdown process, LTB4 is first ω-hydroxylated and then oxidized to a carboxylic acid in human leukocytes. Experiments with 18O demonstrated that the ω-hydroxyl group came from molecular oxygen suggesting the involvement

Isoform- and tissue-specific expression of cytochrome P450 4F subfamily

Maximum expression of several CYP is detected in the liver, as it is the main site for endogenous substrates and drug metabolism. It is, however, important to study CYP in extrahepatic tissues, since they are known to play important roles in organ function as well as having pathophysiological states associated with them (Nebert & Russell, 2002). In the lung, heart and kidney, CYP4F presence is important since P450-mediated AA metabolites are implicated in the regulation of blood pressure (

Genomic characterization and cytochrome P450 4F gene expression

The genomic structures of CYP4F genes have been solved either by traditional library screening or by computer analysis using bioinformatics research tools. The human CYP4F genes CYP4F2, CYP4F3, CYP4F8, CYP4F11, and CYP4F12 are located at chromosome 19p13 and compose a large gene cluster (Kikuta et al., 1998a, Christmas et al., 1999, Bylund et al., 2000, Cui et al., 2000, Bylund et al., 2001). Gene structures of these CYP4F enzymes are similar with each other and their splicing sites are almost

In-vivo and in-vitro regulation of rodent cytochrome P450 4F during inflammation

Although inflammation is an integral part of the host defense mechanism against a variety of pathogens as well as tissue injury, it often produces a myriad of unwanted complications. Immediately after a tissue injury or bacterial infection, PMNL are activated and recruited at the site of inflammation by soluble mediators such as LTB4 causing cell adherence, transcapillary migration and chemotaxis (Springer, 1994, West et al., 2005). The liver form of CYP4F3, which is generated by alternative

Cytochrome P450 4F participation and impact on drug metabolism

CYP have been characteristically termed as a family of drug metabolizing enzymes. This aspect of CYP has been typically attributed to CYP3A which metabolize more than 50% of drugs, followed by CYP2D, CYP1A and CYP2B subfamilies (Gonzalez, 1992, Guengerich, 1999, Nebert and Russell, 2002). Human CYP4F isoforms are known to metabolize endogenous eicosanoids; however recently some 4F members have also gained recognition in metabolizing clinically active drugs.

Concluding remarks

Advances in molecular biology and genomics facilitated the biochemical characterization of individual CYP4F enzymes, which in turn revealed many surprises about an enzyme system once believed to metabolize only LTB4. Today, the biological functions of CYP4F include metabolism of several other pro-inflammatory eicosanoids.

There is a worldwide consensus in the scientific community that acute inflammation followed by a timely resolution plays a fundamental part in the body's response to trauma,

Acknowledgments

Research in the authors' laboratory is supported from grants RO1 MH070054 and RO1 NSO44174 from National Institute of Health and US Department of the Army grant T5 0004268 for Texas Training and Technology against Trauma and Terrorism. The authors are thankful to Dr. Sayeepriyadarshini Anakk, Dr. Yasushi Kikuta and Miss Ying Wang for their helpful discussion during the preparation of this manuscript.

References (272)

  • J. Bylund et al.

    Identification of CYP4F8 in human seminal vesicles as a prominent 19-hydroxylase of prostaglandin endoperoxides

    J Biol Chem

    (2000)
  • J. Bylund et al.

    cDNA cloning and expression of CYP4F12, a novel human cytochrome P450

    Biochem Biophys Res Commun

    (2001)
  • J. Bylund et al.

    Leukotriene B4 omega-side chain hydroxylation by CYP4F5 and CYP4F6

    Arch Biochem Biophys

    (2003)
  • J.H. Capdevila et al.

    Cytochrome P450 and arachidonic acid bioactivation. Molecular and functional properties of the arachidonate monooxygenase

    J Lipid Res

    (2000)
  • C. Cauffiez et al.

    Functional characterization of genetic polymorphisms identified in the human cytochrome P450 4F12 (CYP4F12) promoter region

    Biochem Pharmacol

    (2004)
  • C. Cauffiez et al.

    Human CYP4F12 genetic polymorphism: identification and functional characterization of seven variant allozymes

    Biochem Pharmacol

    (2004)
  • L. Chen et al.

    Identification of a new P450 subfamily, CYP4F1, expressed in rat hepatic tumors

    Arch Biochem Biophys

    (1993)
  • P. Christmas et al.

    Expression of the CYP4F3 gene. tissue-specific splicing and alternative promoters generate high and low K(m) forms of leukotriene B(4) omega-hydroxylase

    J Biol Chem

    (1999)
  • P. Christmas et al.

    Alternative splicing determines the function of CYP4F3 by switching substrate specificity

    J Biol Chem

    (2001)
  • P. Christmas et al.

    Myeloid expression of cytochrome P450 4F3 is determined by a lineage-specific alternative promoter

    J Biol Chem

    (2003)
  • S.S. Chuang et al.

    CYP2U1, a novel human thymus- and brain-specific cytochrome P450, catalyzes omega- and (omega-1)-hydroxylation of fatty acids

    J Biol Chem

    (2004)
  • J. Claria et al.

    Altered biosynthesis of leukotrienes and lipoxins and host defense disorders in patients with cirrhosis and ascites

    Gastroenterology

    (1998)
  • M.J. Coon

    Enzyme ingenuity in biological oxidations: a trail leading to cytochrome p450

    J Biol Chem

    (2002)
  • L.A. Cowart et al.

    The CYP4A isoforms hydroxylate epoxyeicosatrienoic acids to form high affinity peroxisome proliferator-activated receptor ligands

    J Biol Chem

    (2002)
  • X. Cui et al.

    Cloning and characterization of the rat cytochrome P450 4F5 (CYP4F5) gene

    Gene

    (2002)
  • X. Cui et al.

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

    Genomics

    (2000)
  • X. Cui et al.

    Expression of cytochromes P450 4F4 and 4F5 in infection and injury models of inflammation

    Biochim Biophys Acta

    (2003)
  • L. Du et al.

    Epidermal CYP2 family cytochromes P450

    Toxicol Appl Pharmacol

    (2004)
  • R.M. Evans et al.

    PPARs and the complex journey to obesity

    Nat Med

    (2004)
  • X. Fang et al.

    Pathways of epoxyeicosatrienoic acid metabolism in endothelial cells. Implications for the vascular effects of soluble epoxide hydrolase inhibition

    J Biol Chem

    (2001)
  • C. Finta et al.

    Intergenic mRNA molecules resulting from trans-splicing

    J Biol Chem

    (2002)
  • A.J. Fisher et al.

    Enhanced pulmonary inflammation in organ donors following fatal non-traumatic brain injury

    Lancet

    (1999)
  • P. Gervois et al.

    Negative regulation of human fibrinogen gene expression by peroxisome proliferator-activated receptor alpha agonists via inhibition of CCAAT box/enhancer-binding protein beta

    J Biol Chem

    (2001)
  • F.J. Gonzalez

    Human cytochromes P450: problems and prospects

    Trends Pharmacol Sci

    (1992)
  • O. Gotoh

    Substrate recognition sites in cytochrome P450 family 2 (CYP2) proteins inferred from comparative analyses of amino acid and coding nucleotide sequences

    J Biol Chem

    (1992)
  • F.P. Guengerich

    Reactions and significance of cytochrome P-450 enzymes

    J Biol Chem

    (1991)
  • G. Hansson et al.

    Identification and biological activity of novel omega-oxidized metabolites of leukotriene B4 from human leukocytes

    FEBS Lett

    (1981)
  • T.W. Harper et al.

    Metabolism of leukotriene B4 in isolated rat hepatocytes. Identification of a novel 18-carboxy-19,20-dinor leukotriene B4 metabolite

    J Biol Chem

    (1986)
  • T. Hashizume et al.

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

    Biochem Biophys Res Commun

    (2001)
  • U. Hoch et al.

    Structural determination of the substrate specificities and regioselectivities of the rat and human fatty acid omega-hydroxylases

    Arch Biochem Biophys

    (2000)
  • S. Imaoka et al.

    Hepatic and renal cytochrome P-450s in spontaneously hypertensive rats

    Biochim Biophys Acta

    (1991)
  • S. Anakk et al.

    Genomic characterization and regulation of CYP3a13: role of xenobiotics and nuclear receptors

    FASEB J

    (2003)
  • H.K. Anandatheerthavarada et al.

    Dual targeting of cytochrome P4502B1 to endoplasmic reticulum and mitochondria involves a novel signal activation by cyclic AMP-dependent phosphorylation at ser128

    EMBO J

    (1999)
  • P. Anzenbacher et al.

    Cytochromes P450 and metabolism of xenobiotics

    Cell Mol Life Sci

    (2001)
  • B.R. Baer et al.

    Sites of covalent attachment of CYP4 enzymes to heme: evidence for microheterogeneity of P450 heme orientation

    Biochemistry

    (2005)
  • M.B. Bailie et al.

    Leukotriene-deficient mice manifest enhanced lethality from Klebsiella pneumonia in association with decreased alveolar macrophage phagocytic and bactericidal activities

    J Immunol

    (1996)
  • A.S. Baldwin

    The NF-kappa B and I kappa B proteins: new discoveries and insights

    Annu Rev Immunol

    (1996)
  • P.J. Barnes et al.

    Inflammatory mediators of asthma: an update

    Pharmacol Rev

    (1998)
  • W.H. Beierwaltes et al.

    Electrolyte and water balance in young spontaneously hypertensive rats

    Hypertension

    (1982)
  • M. Bhattacharya et al.

    Nuclear localization of prostaglandin E2 receptors

    Proc Natl Acad Sci U S A

    (1998)
  • Cited by (118)

    • Fluorescent probes for the detection and imaging of Cytochrome P450

      2021, Coordination Chemistry Reviews
      Citation Excerpt :

      Probe 18 facilitates the sensitive detection of CYP4A11 in a variety of biospecimens and helps contribute to an understanding of the involvement of CYP4A11 in human diseases. The CYP4F subfamily consists of CYP4F11, CYP4F12, CYP4F2, CYP4F3A, CYP4F3B, and CYP4F8, which are all involved in the biotransformation of eicosanoids such as arachidonic acid, prostaglandin, and leukotriene B4 (LTB4) and play a crucial role in the body's inflammatory response and blood pressure regulation [99–101]. Among them, CYP4F12 and 4F3B are mostly expressed in the liver and kidneys.

    View all citing articles on Scopus
    View full text