Analysis of Differential Substrate Selectivities of CYP2B6 and CYP2E1 by Site-Directed Mutagenesis and Molecular Modeling

Human CYP2B6 and CYP2E1 were used to investigate the extent to which differential substrate selectivities between cytochrome P450 subfamilies reflect differences in active-site residues as opposed to distinct arrangement of the backbone of the enzymes. Reciprocal CYP2B6 and CYP2E1 mutants at active-site positions 103, 209, 294, 363, 367, and 477 (numbering according to CYP2B6) were characterized using the CYP2B6-selective substrate 7-ethoxy-4-trifluoromethylcoumarin, the CYP2E1-selective substrate p-nitrophenol, and the common substrates 7-ethoxycoumarin, 7-butoxycoumarin, and arachidonic acid. This report is the first to study the active site of CYP2E1 by systematic site-directed mutagenesis. One of the most intriguing findings was that substitution of CYP2E1 Phe-477 with valine from CYP2B6 resulted in significant 7-ethoxy-4-trifluoromethylcoumarin deethylation. Use of three-dimensional models of CYP2B6 and CYP2E1 based on the crystal structure of CYP2C5 suggested that deethylation of 7-ethoxy-4-trifluoromethylcoumarin by CYP2E1 is impeded by van der Waals overlaps with the side chain of Phe-477. Interestingly, none of the CYP2B6 mutants acquired enhanced ability to hydroxylatep-nitrophenol. Substitution of residue 363 in CYP2E1 and CYP2B6 resulted in significant alterations of the metabolite profile for the side chain hydroxylation of 7-butoxycoumarin. Probing of CYP2E1 mutants with arachidonic acid indicated that residues Leu-209 and Phe-477 are critical for substrate orientation in the active site. Overall, the study revealed that differences in the side chains of active-site residues are partially responsible for differential substrate selectivities across cytochrome P450 subfamilies. However, the relative importance of active-site residues appears to be dependent on the structural similarity of the compound to other substrates of the enzyme.

[1]  J. Halpert,et al.  A truncation of 2B subfamily cytochromes P450 yields increased expression levels, increased solubility, and decreased aggregation while retaining function. , 2001, Archives of biochemistry and biophysics.

[2]  J. Halpert,et al.  The role of cytochrome 2B1 substrate recognition site residues 115, 294, 297, 298, and 362 in the oxidation of steroids and 7-alkoxycoumarins. , 2001, Archives of biochemistry and biophysics.

[3]  T. Goosen,et al.  Differential effects of naturally occurring isothiocyanates on the activities of cytochrome P450 2E1 and the mutant P450 2E1 T303A. , 2001, Archives of biochemistry and biophysics.

[4]  J. Halpert,et al.  Analysis of mammalian cytochrome P450 structure and function by site-directed mutagenesis. , 2001, Current drug metabolism.

[5]  C. Masimirembwa,et al.  Competitive CYP2C9 inhibitors: enzyme inhibition studies, protein homology modeling, and three-dimensional quantitative structure-activity relationship analysis. , 2001, Molecular pharmacology.

[6]  J. Halpert,et al.  Amino acid residues critical for differential inhibition of CYP2B4, CYP2B5, and CYP2B1 by phenylimidazoles. , 2001, Molecular pharmacology.

[7]  C. Lindley,et al.  Validation of bupropion hydroxylation as a selective marker of human cytochrome P450 2B6 catalytic activity. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[8]  D E McRee,et al.  Microsomal cytochrome P450 2C5: comparison to microbial P450s and unique features. , 2000, Journal of inorganic biochemistry.

[9]  A. P. Koley,et al.  Conformational modulation of human cytochrome P450 2E1 by ethanol and other substrates: a CO flash photolysis study. , 2000, Biochemistry.

[10]  J. Salaün,et al.  Requirement for omega and (omega;-1)-hydroxylations of fatty acids by human cytochromes P450 2E1 and 4A11. , 1999, Journal of lipid research.

[11]  J. Halpert,et al.  Structure-function analysis of human cytochrome P-450 2B6 using a novel substrate, site-directed mutagenesis, and molecular modeling. , 1999, The Journal of pharmacology and experimental therapeutics.

[12]  T. Macgregor,et al.  Disposition and biotransformation of the antiretroviral drug nevirapine in humans. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[13]  D. Waxman,et al.  Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[14]  D. Stresser,et al.  Monospecific antipeptide antibody to cytochrome P-450 2B6. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[15]  J. Halpert,et al.  Probing the active site of cytochrome P450 2B1: metabolism of 7-alkoxycoumarins by the wild type and five site-directed mutants. , 1998, Biochemistry.

[16]  M. Pincus,et al.  Molecular Modeling of Cytochrome P450 2B1: Mode of Membrane Insertion and Substrate Specificity , 1998, Journal of protein chemistry.

[17]  T. Aoyama,et al.  Cytochrome P450 isozymes responsible for the metabolism of toluene and styrene in human liver microsomes. , 1997, Xenobiotica; the fate of foreign compounds in biological systems.

[18]  J. Halpert,et al.  Functional interaction between amino-acid residues 242 and 290 in cytochromes P-450 2B1 and 2B11. , 1997, Biochimica et biophysica acta.

[19]  Slobodan Petar Rendic,et al.  Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. , 1997, Drug metabolism reviews.

[20]  J. Halpert,et al.  Interconversion of the androstenedione hydroxylase specificities of cytochromes P450 2B4 and 2B5 upon simultaneous site-directed mutagenesis of four key substrate recognition residues. , 1996, Archives of biochemistry and biophysics.

[21]  J. Stevens,et al.  Catalytic role of cytochrome P4502B6 in the N-demethylation of S-mephenytoin. , 1996, Drug metabolism and disposition: the biological fate of chemicals.

[22]  P. Ortiz de Montellano,et al.  Active site topology of human cytochrome P450 2E1. , 1996, Chemical research in toxicology.

[23]  M. Waterman,et al.  Genetically engineered bacterial cells and applications. , 1995, Toxicology letters.

[24]  J. Meinwald,et al.  Stereoselective metabolism of enflurane by human liver cytochrome P450 2E1. , 1995, Drug metabolism and disposition: the biological fate of chemicals.

[25]  J. Halpert,et al.  Site-directed mutagenesis as a tool for molecular modeling of cytochrome P450 2B1. , 1995, Biochemistry.

[26]  J. Halpert,et al.  Escherichia coli expression of site-directed mutants of cytochrome P450 2B1 from six substrate recognition sites: substrate specificity and inhibitor selectivity studies. , 1995, Chemical research in toxicology.

[27]  D. Wade,et al.  Probing the active sites of rat and human cytochrome P450 2E1 with alcohols and carboxylic acids. , 1995, Archives of biochemistry and biophysics.

[28]  J. Goldstein,et al.  Biochemistry and molecular biology of the human CYP2C subfamily. , 1994, Pharmacogenetics.

[29]  J. Halpert,et al.  Role of Residue 363 and 206 in Conversion of Cytochrome P450 2B1 from a Steroid 16-Hydroxylase to a 15α-Hydroxylase , 1994 .

[30]  D. Koop,et al.  Formation of 19(S)-, 19(R)-, and 18(R)-hydroxyeicosatetraenoic acids by alcohol-inducible cytochrome P450 2E1. , 1993, The Journal of biological chemistry.

[31]  Mark D. Paulsen,et al.  Predicting the product specificity and coupling of cytochrome P450cam , 1992, J. Comput. Aided Mol. Des..

[32]  David F. V. Lewis,et al.  The sequence homologies of cytochromes P-450 and active-site geometries , 1992, J. Comput. Aided Mol. Des..

[33]  D. Koop Oxidative and reductive metabolism by cytochrome P450 2E1 , 1992, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[34]  R. Ornstein,et al.  A 175‐psec molecular dynamics simulation of camphor‐bound cytochrome P‐450cam , 1991, Proteins.

[35]  A. Cederbaum,et al.  Increased oxidation of p-nitrophenol and aniline by intact hepatocytes isolated from pyrazole-treated rats. , 1990, Biochimica et biophysica acta.

[36]  C. Levinthal,et al.  Predicting antibody hypervariable loop conformation. I. Ensembles of random conformations for ringlike structures , 1987, Biopolymers.

[37]  T. Omura,et al.  THE CARBON MONOXIDE-BINDING PIGMENT OF LIVER MICROSOMES. I. EVIDENCE FOR ITS HEMOPROTEIN NATURE. , 1964, The Journal of biological chemistry.

[38]  T. Omura,et al.  THE CARBON MONOXIDE-BINDING PIGMENT OF LIVER MICROSOMES. II. SOLUBILIZATION, PURIFICATION, AND PROPERTIES. , 1964, The Journal of biological chemistry.

[39]  J. Halpert,et al.  Combined three-dimensional quantitative structure-activity relationship analysis of cytochrome P450 2B6 substrates and protein homology modeling. , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[40]  J. Salaün,et al.  Requirement for v and ( v – 1 )-hydroxylations of fatty acids by human cytochromes P 450 2 E 1 and 4 A 11 , 1999 .

[41]  T. Darden,et al.  The roles of individual amino acids in altering substrate specificity of the P450 2a4/2a5 enzymes. , 1996, Biochimie.

[42]  D. Koop,et al.  The utility of p-nitrophenol hydroxylation in P450IIE1 analysis. , 1989, Drug metabolism reviews.

[43]  David M. Kramer,et al.  Biochemistry and Molecular Biology , 1968, Nature.