Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 38(2): 347-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923256

ABSTRACT

Several antihistamine drugs including terfenadine, ebastine, and astemizole have been identified as substrates for CYP2J2. The overall importance of this enzyme in drug metabolism has not been fully explored. In this study, 139 marketed therapeutic agents and compounds were screened as potential CYP2J2 substrates. Eight novel substrates were identified that vary in size and overall topology from relatively rigid structures (amiodarone) to larger complex structures (cyclosporine). The substrates displayed in vitro intrinsic clearance values ranging from 0.06 to 3.98 mul/min/pmol CYP2J2. Substrates identified for CYP2J2 are also metabolized by CYP3A4. Extracted ion chromatograms of metabolites observed for albendazole, amiodarone, astemizole, thioridazine, mesoridazine, and danazol showed marked differences in the regioselectivity of CYP2J2 and CYP3A4. CYP3A4 commonly metabolized compounds at multiple sites, whereas CYP2J2 metabolism was more restrictive and limited, in general, to a single site for large compounds. Although the CYP2J2 active site can accommodate large substrates, it may be more narrow than CYP3A4, limiting metabolism to moieties that can extend closer toward the active heme iron. For albendazole, CYP2J2 forms a unique metabolite compared with CYP3A4. Albendazole and amiodarone were evaluated in various in vitro systems including recombinant CYP2J2 and CYP3A4, pooled human liver microsomes (HLM), and human intestinal microsomes (HIM). The Michaelis-Menten-derived intrinsic clearance of N-desethyl amiodarone was 4.6 greater in HLM than in HIM and 17-fold greater in recombinant CYP3A4 than in recombinant CYP2J2. The resulting data suggest that CYP2J2 may be an unrecognized participant in first-pass metabolism, but its contribution is minor relative to that of CYP3A4.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Pharmaceutical Preparations/metabolism , Algorithms , Catalytic Domain , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2J2 , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/chemistry , Enzyme Inhibitors/pharmacology , Humans , Intestinal Mucosa/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Liver/metabolism , Microsomes/metabolism , Models, Structural , Organ Specificity , Pharmaceutical Preparations/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity , Tandem Mass Spectrometry
2.
Drug Metab Dispos ; 36(3): 490-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18096676

ABSTRACT

Carbamazepine (CBZ) is a widely prescribed anticonvulsant whose use is often associated with idiosyncratic hypersensitivity. Sera of CBZ-hypersensitive patients often contain anti-CYP3A antibodies, including those to a CYP3A23 K-helix peptide that is also modified during peroxidative CYP3A4 heme-fragmentation. We explored the possibility that cytochromes P450 (P450s) such as CYP3A4 bioactivate CBZ to reactive metabolite(s) that irreversibly modify the P450 protein. Such CBZ-P450 adducts, if stable in vivo, could engender corresponding serum P450 autoantibodies. Incubation with CBZ not only failed to inactivate functionally reconstituted, purified recombinant CYP3A4 or CYP3A4 Supersomes in a time-dependent manner, but the inclusion of CBZ (0-1 mM) also afforded a concentration-dependent protection to CYP3A4 from inactivation by NADPH-induced oxidative uncoupling. Incubation of CYP3A4 Supersomes with (3)H-CBZ resulted in its irreversible binding to CYP3A4 protein with a stoichiometry of 1.58 +/- 0.15 pmol (3)H-CBZ bound/pmol CYP3A4. Inclusion of glutathione (1.5 mM) in the incubation reduced this level to 1.09. Similar binding (1.0 +/- 0.4 pmol (3)H-CBZ bound/pmol CYP3A4) was observed after (3)H-CBZ incubation with functionally reconstituted, purified recombinant CYP3A4(His)(6). The CBZ-modified CYP3A4 retained its functional activity albeit at a reduced level, but its testosterone 6beta-hydroxylase kinetics were altered from sigmoidal (a characteristic profile of substrate cooperativity) to near-hyperbolic (Michaelis-Menten) type, suggesting that CBZ may have modified CYP3A4 within its active site.


Subject(s)
Carbamazepine/metabolism , Cytochrome P-450 CYP3A/metabolism , Carbamazepine/chemistry , Carbamazepine/pharmacology , Crystallography, X-Ray , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Enzyme Activation/drug effects , Erythromycin/chemistry , Erythromycin/metabolism , Humans , Kinetics , Methylation , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Steroid Hydroxylases/chemistry , Steroid Hydroxylases/metabolism , Testosterone/chemistry , Testosterone/metabolism
3.
J Biol Chem ; 279(37): 38091-4, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15258162

ABSTRACT

The structure of P450 3A4 was determined by x-ray crystallography to 2.05-A resolution. P450 3A4 catalyzes the metabolic clearance of a large number of clinically used drugs, and a number of adverse drug-drug interactions reflect the inhibition or induction of the enzyme. P450 3A4 exhibits a relatively large substrate-binding cavity that is consistent with its capacity to oxidize bulky substrates such as cyclosporin, statins, taxanes, and macrolide antibiotics. Family 3A P450s also exhibit unusual kinetic characteristics that suggest simultaneous occupancy by smaller substrates. Although the active site volume is similar to that of P450 2C8 (PDB code: 1PQ2), the shape of the active site cavity differs considerably due to differences in the folding and packing of portions of the protein that form the cavity. Compared with P450 2C8, the active site cavity of 3A4 is much larger near the heme iron. The lower constraints on the motions of small substrates near the site of oxygen activation may diminish the efficiency of substrate oxidation, which may, in turn, be improved by space restrictions imposed by the presence of a second substrate molecule. The structure of P450 3A4 should facilitate a better understanding of the substrate selectivity of the enzyme.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Microsomes/enzymology , Arginine/chemistry , Binding Sites , Crystallography, X-Ray , Cytochrome P-450 CYP3A , Heme/chemistry , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity
4.
J Biol Chem ; 279(34): 35630-7, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15181000

ABSTRACT

The structure of human P450 2C9 complexed with flurbiprofen was determined to 2.0 A by x-ray crystallography. In contrast to other structurally characterized P450 2C enzymes, 2C5, 2C8, and a 2C9 chimera, the native catalytic domain of P450 2C9 differs significantly in the conformation of the helix F to helix G region and exhibits an extra turn at the N terminus of helix A. In addition, a distinct conformation of the helix B to helix C region allows Arg-108 to hydrogen bond with Asp-293 and Asn-289 on helix I and to interact directly with the carboxylate of flurbiprofen. These interactions position the substrate for regioselective oxidation in a relatively large active site cavity and are likely to account for the high catalytic efficiency exhibited by P450 2C9 for the regioselective oxidation of several anionic non-steroidal anti-inflammatory drugs. The structure provides a basis for interpretation of a number of observations regarding the substrate selectivity of P450 2C9 and the observed effects of mutations on catalysis.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Flurbiprofen/chemistry , Amino Acid Sequence , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Binding Sites/genetics , Catalytic Domain , Cytochrome P-450 CYP2C9 , Flurbiprofen/metabolism , Humans , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Substrate Specificity
5.
J Biol Chem ; 279(10): 9497-503, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14676196

ABSTRACT

A 2.7-Angstrom molecular structure of human microsomal cytochrome P450 2C8 (CYP2C8) was determined by x-ray crystallography. The membrane protein was modified for crystallization by replacement of the hydrophobic N-terminal transmembrane domain with a short hydrophilic sequence before residue 28. The structure of the native sequence is complete from residue 28 to the beginning of a C-terminal histidine tag used for purification. CYP2C8 is one of the principal hepatic drug-metabolizing enzymes that oxidizes therapeutic drugs such as taxol and cerivastatin and endobiotics such as retinoic acid and arachidonic acid. Consistent with the relatively large size of its preferred substrates, the active site volume is twice that observed for the structure of CYP2C5. The extended active site cavity is bounded by the beta1 sheet and helix F' that have not previously been implicated in substrate recognition by mammalian P450s. CYP2C8 crystallized as a symmetric dimer formed by the interaction of helices F, F', G', and G. Two molecules of palmitic acid are bound in the dimer interface. The dimer is observed in solution, and mass spectrometry confirmed the association of palmitic acid with the enzyme. This novel finding identifies a peripheral binding site in P450s that may contribute to drug-drug interactions in P450 metabolism.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/metabolism , Binding Sites , Cytochrome P-450 CYP2C8 , Dimerization , Fatty Acids/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation
6.
Proc Natl Acad Sci U S A ; 100(23): 13196-201, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14563924

ABSTRACT

The xenobiotic metabolizing cytochromes P450 (P450s) are among the most versatile biological catalysts known, but knowledge of the structural basis for their broad substrate specificity has been limited. P450 2B4 has been frequently used as an experimental model for biochemical and biophysical studies of these membrane proteins. A 1.6-A crystal structure of P450 2B4 reveals a large open cleft that extends from the protein surface directly to the heme iron between the alpha-helical and beta-sheet domains without perturbing the overall P450 fold. This cleft is primarily formed by helices B' to C and F to G. The conformation of these regions is dramatically different from that of the other structurally defined mammalian P450, 2C5/3LVdH, in which the F to G and B' to C regions encapsulate one side of the active site to produce a closed form of the enzyme. The open conformation of 2B4 is trapped by reversible formation of a homodimer in which the residues between helices F and G of one molecule partially fill the open cleft of a symmetry-related molecule, and an intermolecular coordinate bond occurs between H226 and the heme iron. This dimer is observed both in solution and in the crystal. Differences between the structures of 2C5 and 2B4 suggest that defined regions of xenobiotic metabolizing P450s may adopt a substantial range of energetically accessible conformations without perturbing the overall fold. This conformational flexibility is likely to facilitate substrate access, metabolic versatility, and product egress.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Animals , Bacterial Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Cytochrome P450 Family 2 , Dimerization , Heme/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Rabbits , Ultracentrifugation , Xenobiotics/metabolism
7.
Biochemistry ; 42(31): 9335-45, 2003 Aug 12.
Article in English | MEDLINE | ID: mdl-12899620

ABSTRACT

The structure of the anti-inflammatory drug diclofenac bound in the active site of rabbit microsomal cytochrome P450 2C5/3LVdH was determined by X-ray crystallography to 2.1 A resolution. P450 2C5/3LVdH and the related enzyme 2C5dH catalyze the 4'-hydroxylation of diclofenac with apparent K(m) values of 80 and 57 microM and k(cat) values of 13 and 16 min(-1), respectively. Spectrally determined binding constants are similar to the K(m) values. The structure indicates that the pi-electron system of the dichlorophenyl moiety faces the heme Fe with the 3'- and 4'-carbons located 4.4 and 4.7 A, respectively, from the Fe. The carboxyl moiety of the substrate is hydrogen bonded to a cluster of waters that are also hydrogen bonded to the side chains of N204, K241, S289, and D290 as well as the backbone of the protein. The proximity of the diclofenac carboxylate to the side chain of D290 together with an increased binding affinity at lower pH suggests that diclofenac is protonated when bound to the enzyme. The structure exhibits conformational changes indicative of an adaptive fit to the substrate reflecting both the hydration and size of the substrate. These results indicate how structurally diverse substrates are recognized by drug-metabolizing P450 enzymes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/chemistry , Diclofenac/metabolism , Steroid 21-Hydroxylase/chemistry , Steroid 21-Hydroxylase/metabolism , Animals , Carboxylic Acids/chemistry , Carboxylic Acids/metabolism , Catalysis , Crystallography, X-Ray , Cytochrome P450 Family 2 , Heme/chemistry , Iron/chemistry , Microsomes/enzymology , Models, Molecular , Protein Binding , Protein Conformation , Protein Folding , Rabbits , Structure-Activity Relationship , Substrate Specificity , Water/chemistry
8.
Biochemistry ; 42(21): 6370-9, 2003 Jun 03.
Article in English | MEDLINE | ID: mdl-12767218

ABSTRACT

The structure of rabbit microsomal cytochrome P450 2C5/3LVdH complexed with a substrate, 4-methyl-N-methyl-N-(2-phenyl-2H-pyrazol-3-yl)benzenesulfonamide (DMZ), was determined by X-ray crystallography to 2.3 A resolution. Substrate docking studies and electron density maps indicate that DMZ binds to the enzyme in two antiparallel orientations of the long axis of the substrate. One orientation places the principal site of hydroxylation, the 4-methyl group, 4.4 A from the heme Fe, whereas the alternate conformation positions the second, infrequent site of hydroxylation at >5.9 A from the heme Fe. Comparison of this structure to that obtained previously for the enzyme indicates that the protein closes around the substrate and prevents open access of water from bulk solvent to the heme Fe. This reflects a approximately 1.5 A movement of the F and G helices relative to helix I. The present structure provides a complete model for the protein from residues 27-488 and defines two new helices F' and G'. The G' helix is likely to contribute to interactions of the enzyme with membranes. The relatively large active site, as compared to the volume occupied by the substrate, and the flexibility of the enzyme are likely to underlie the capacity of drug-metabolizing enzymes to metabolize structurally diverse substrates of different sizes.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Imidazoles/chemistry , Steroid 21-Hydroxylase/chemistry , Crystallography, X-Ray , Cytochrome P450 Family 2 , Electrons , Heme/chemistry , Humans , Iron/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Water/chemistry
9.
Biochemistry ; 42(21): 6363-9, 2003 Jun 03.
Article in English | MEDLINE | ID: mdl-12767217

ABSTRACT

The inhibitory effects of a series of sulfaphenazole (SPA) derivatives were studied on two modified forms of rabbit liver cytochrome P450 2C5 (CYP2C5), CYP2C5dH, and structurally characterized CYP2C5/3LVdH and compared to the previously described effects of these compounds on human CYP2C8, 2C9, 2C18, and 2C19. SPA and other negatively charged compounds that potently inhibit CYP2C9 had very little effect on CYP2C5dH, whereas neutral, N-alkylated derivatives exhibited IC50 values between 8 and 22 microM. One of the studied compounds, 4, that derives from SPA by replacement of its NH(2) substituent with a methyl group and by N-methylation of its sulfonamide moiety, acted as a good substrate for all CYP2Cs used in this study. Hydroxylation of the benzylic methyl of 4 is the major reaction catalyzed by all of these CYP2C proteins, whereas hydroxylation of the N-phenyl group of 4 was observed as a minor reaction. CYP2C5dH, 2C5/3LVdH, 2C9, 2C18, and 2C19 are efficient catalysts for the benzylic hydroxylation of 4, with K(m) values between 5 and 13 microM and k(cat) values between 16 and 90 min(-1). The regioselectivity observed for oxidation of 4 by CYP2C5/3LVdH was easily interpreted on the basis of the existence of two different binding modes of 4 characterized in the experimentally determined structure of the complexes of CYP2C5/3LVdH with 4 described in the following paper [Wester, M. R. et al. (2003) Biochemistry 42, 6370-6379].


Subject(s)
Biochemistry/methods , Cytochrome P-450 Enzyme System/chemistry , Steroid 21-Hydroxylase/chemistry , Sulfaphenazole/pharmacology , Anti-Infective Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/chemistry , Cells, Cultured , Crystallography, X-Ray , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P450 Family 2 , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Kinetics , Liver/metabolism , Microsomes/metabolism , Mixed Function Oxygenases/chemistry , Models, Chemical , Oxygen/metabolism , Progesterone/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , Substrate Specificity , Sulfaphenazole/chemistry , Sulfonamides/chemistry , Ultraviolet Rays
10.
Methods Enzymol ; 357: 73-9, 2002.
Article in English | MEDLINE | ID: mdl-12424899

ABSTRACT

Engineering more soluble forms of P450 2C5 has contributed to the crystallization of the enzyme. When detergents are used in both crystallization and purification of the protein, the ability to control the content and identity of the detergent is dependent on the protein exhibiting a sufficient degree of solubility to permit its concentration in the absence of detergents. The production of concentrated solutions of the protein containing little or no detergent provides a means for screening crystallization conditions and the selection of detergents that facilitate crystallization. These detergents can then be used not only to improve the purification of the protein, but also to solublize substrates for the cocrystallization of enzyme-substrate complexes.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/isolation & purification , Detergents/chemistry , Microsomes/enzymology , Steroid 21-Hydroxylase/chemistry , Steroid 21-Hydroxylase/isolation & purification , Animals , Cryoprotective Agents/chemistry , Crystallization , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 2 , Rabbits , Steroid 21-Hydroxylase/genetics , X-Ray Diffraction
11.
Endocr Res ; 28(4): 435-41, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12530646

ABSTRACT

The structure of microsomal P450 2C5 is the first structure of a membrane P450 to be determined by x-ray diffraction. This enzyme was originally identified as a progesterone 21-hydroxylase that is polymorphically expressed in rabbit liver. In contrast to the adrenal 21-hydroxylase, P450 2C5 metabolizes structurally diverse substrates that include a variety of steroids as well as therapeutic drugs. The flexible architecture of the enzyme and the residual solvation of the substrate provide a basis for understanding the catalytic diversity of 2C5 and related drug metabolizing P450s. In addition, the structure of P450 2C5 suggests how mammalian P450s have adapted for membrane binding and interaction with microsomal P450 reductase.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Microsomes/metabolism , Steroid 21-Hydroxylase/chemistry , Animals , Catalysis , Crystallization , Crystallography , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2 , Humans , Pharmaceutical Preparations/metabolism , Protein Folding , Protein Structure, Secondary , Steroid 21-Hydroxylase/metabolism , Steroids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...