1
|
Campomizzi CS, Uttamrao PP, Stallone JJ, Rathinavelan T, Estrada DF. Asparagine-85 Stabilizes a Structural Active Site Water Network in CYP121A1 of Mycobacterium tuberculosis. Biochemistry 2024; 63:711-722. [PMID: 38380587 DOI: 10.1021/acs.biochem.3c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The cytochrome P450 enzyme CYP121A1 endogenously catalyzes the formation of a carbon-carbon bond between the two phenol groups of dicyclotyrosine (cYY) in Mycobacterium tuberculosis (Mtb). One of 20 CYP enzymes in Mtb, CYP121A1 continues to garner significant interest as a potential drug target. The accompanying reports the use of 19F NMR spectroscopy, reconstituted activity assays, and molecular dynamics simulations to investigate the significance of hydrogen bonding interactions that were theorized to stabilize a static active site water network. The active site residue Asn-85, whose hydrogen bonds with the diketopiperazine ring of cYY contributes to a contiguous active site water network in the absence of cYY, was mutated to a serine (N85S) and to a glutamine (N85Q). These conservative changes in the hydrogen bond donor side chain result in inactivation of the enzyme. Moreover, the N85S mutation induces reverse type-I binding as measured by absorbance difference spectra. NMR spectra monitoring the ligand-adaptive FG-loop and the active site Trp-182 side chain confirm that disruption of the active site water network also significantly alters the structure of the active site. These data were consistent with dynamics simulations of N85S and N85Q that demonstrate that a compromised water network is responsible for remodeling of the active site B-helix and a repositioning of cYY toward the heme. These findings implicate a slowly exchanging water network as a critical factor in CYP121A1 function and a likely contributor to the unusual rigidity of the structure.
Collapse
Affiliation(s)
- Christopher S Campomizzi
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Science, University at Buffalo, Buffalo, New York 14203, United States
| | - Patil Pranita Uttamrao
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Jack J Stallone
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Science, University at Buffalo, Buffalo, New York 14203, United States
| | - Thenmalarchelvi Rathinavelan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - D Fernando Estrada
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Science, University at Buffalo, Buffalo, New York 14203, United States
| |
Collapse
|
2
|
Carstensen L, Beil S, Schwab E, Banke S, Börnick H, Stolte S. Primary and ultimate degradation of benzophenone-type UV filters under different environmental conditions and the underlying structure-biodegradability relationships. JOURNAL OF HAZARDOUS MATERIALS 2023; 446:130634. [PMID: 36599278 DOI: 10.1016/j.jhazmat.2022.130634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/17/2022] [Indexed: 06/17/2023]
Abstract
Ten common benzophenone-based UV filters (BPs), sharing the same basic structure and differing only in their substituents, were investigated with respect to their primary and ultimate biodegradability. This study was carried out in order to gain deeper insights into the relationship between structure and biodegradability. The primary biodegradation of the selected BPs was studied in river water at environmentally relevant concentrations (1 µg/L) while varying specific, crucial environmental conditions (aerobic, suboxic, supplementation of nutrients). For this purpose, both batch and column degradation tests were performed, which allowed a systematic study of the effects. Subsequently, the ultimate biodegradation, i.e. the potential to achieve full mineralization of BPs, was examined according to OECD guideline 301 F. The results indicate that mineralization is limited to derivatives in which both aromatic rings contain substituents. This hypothesis was supported by docking simulations showing systematic differences in the orientation of BPs within the active site of the cytochrome P450 enzyme. These differences in orientation correspond to the substitution pattern of the BPs. This study provides valuable insights for assessing the environmental hazards of this class of trace organic compounds.
Collapse
Affiliation(s)
- Lale Carstensen
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany
| | - Stephan Beil
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany
| | - Ekaterina Schwab
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany
| | - Sophie Banke
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany
| | - Hilmar Börnick
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany
| | - Stefan Stolte
- Institute of Water Chemistry, Technical University of Dresden, 01069 Dresden, Germany.
| |
Collapse
|
3
|
Liu G, Zhao Z, Li M, Zhao M, Xu T, Wang S, Zhang Y. Current perspectives on benzoflavone analogues with potent biological activities: A review. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
4
|
Díaz-Storani L, Clary AA, Moreno DM, Ballari MS, Porta EOJ, Bracca ABJ, Johnston JB, Labadie GR. Synthesis and interaction of terminal unsaturated chemical probes with Mycobacterium tuberculosis CYP124A1. Bioorg Med Chem 2021; 44:116304. [PMID: 34289431 DOI: 10.1016/j.bmc.2021.116304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 11/29/2022]
Abstract
A series of C15-C20 isoprenyl derivatives bearing terminal alkenyl and alkynyl groups were synthesized as possible substrates of the methyl-branched lipid ω-hydroxylase CYP124A1 from Mycobacterium tuberculosis. The interactions of each compound with the enzyme active site were characterized using UV-vis spectroscopy. We found that C10 and C15 analogs bind with similar affinity to the corresponding parent C10 and C15 substrates geraniol and farnesol, respectively. Three analogs (C10-ω-ene, C10-ω-yne, C15-ω-yne) interact with the proximal side of the heme iron by coordinating to the oxygen atom of the ferric heme, as judged by the appearance of typical Type-IA binding spectra. On the other hand, the C15-ω-ene analog interacts with the ferric heme by displacing the bound water that generates a typical Type I binding spectrum. We were unable to detect P450-mediated oxidation of these probes following extended incubations with CYP124A1 in our reconstituted assay system, whereas a control reaction containing farnesol was converted to ω-hydroxy farnesol under the same conditions. To understand the lack of detectable oxidation, we explored the possibility that the analogs were acting as mechanism-based inhibitors, but we were unable to detect time-dependent loss of enzymatic activity. In order to gain insight into the lack of detectable turnover or time-dependent inhibition, we examined the interaction of each compound with the CYP124A1 active site using molecular docking simulations. The docking studies revealed a binding mode where the terminal unsaturated functional groups were sequestered within the methyl-binding pocket, rather than positioned close to the heme iron for oxidation. These results aid in the design of specific inhibitors of Mtb-CYP124A1, an interesting enzyme that is implicated in the oxidation of methyl-branched lipids, including cholesterol, within a deadly human pathogen.
Collapse
Affiliation(s)
- Luz Díaz-Storani
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Anaelle A Clary
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158-2517, United States
| | - Diego M Moreno
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - María Sol Ballari
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Exequiel O J Porta
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Andrea B J Bracca
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Jonathan B Johnston
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158-2517, United States.
| | - Guillermo R Labadie
- Instituto de Química Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK Rosario, Argentina.
| |
Collapse
|
5
|
Kaluzhskiy L, Ershov P, Yablokov E, Shkel T, Grabovec I, Mezentsev Y, Gnedenko O, Usanov S, Shabunya P, Fatykhava S, Popov A, Artyukov A, Styshova O, Gilep A, Strushkevich N, Ivanov A. Human Lanosterol 14-Alpha Demethylase (CYP51A1) Is a Putative Target for Natural Flavonoid Luteolin 7,3'-Disulfate. Molecules 2021; 26:2237. [PMID: 33924405 PMCID: PMC8070018 DOI: 10.3390/molecules26082237] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Widespread pathologies such as atherosclerosis, metabolic syndrome and cancer are associated with dysregulation of sterol biosynthesis and metabolism. Cholesterol modulates the signaling pathways of neoplastic transformation and tumor progression. Lanosterol 14-alpha demethylase (cytochrome P450(51), CYP51A1) catalyzes one of the key steps in cholesterol biosynthesis. The fairly low somatic mutation frequency of CYP51A1, its druggability, as well as the possibility of interfering with cholesterol metabolism in cancer cells collectively suggest the clinical importance of CYP51A1. Here, we show that the natural flavonoid, luteolin 7,3'-disulfate, inhibits CYP51A1 activity. We also screened baicalein and luteolin, known to have antitumor activities and low toxicity, for their ability to interact with CYP51A1. The Kd values were estimated using both a surface plasmon resonance optical biosensor and spectral titration assays. Unexpectedly, in the enzymatic activity assays, only the water-soluble form of luteolin-luteolin 7,3'-disulfate-showed the ability to potently inhibit CYP51A1. Based on molecular docking, luteolin 7,3'-disulfate binding suggests blocking of the substrate access channel. However, an alternative site on the proximal surface where the redox partner binds cannot be excluded. Overall, flavonoids have the potential to inhibit the activity of human CYP51A1 and should be further explored for their cholesterol-lowering and anti-cancer activity.
Collapse
Affiliation(s)
- Leonid Kaluzhskiy
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| | - Pavel Ershov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| | - Evgeniy Yablokov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| | - Tatsiana Shkel
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Irina Grabovec
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Yuri Mezentsev
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| | - Oksana Gnedenko
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| | - Sergey Usanov
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Polina Shabunya
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Sviatlana Fatykhava
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Alexander Popov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, 690022 Vladivostok, Russia; (A.P.); (A.A.); (O.S.)
| | - Aleksandr Artyukov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, 690022 Vladivostok, Russia; (A.P.); (A.A.); (O.S.)
| | - Olga Styshova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, 690022 Vladivostok, Russia; (A.P.); (A.A.); (O.S.)
| | - Andrei Gilep
- Institute of Bioorganic Chemistry NASB, 5 Building 2, V.F. Kuprevich Street, 220141 Minsk, Belarus; (T.S.); (I.G.); (S.U.); (P.S.); (S.F.); (A.G.)
| | - Natallia Strushkevich
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
| | - Alexis Ivanov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia; (P.E.); (E.Y.); (Y.M.); (O.G.); (A.I.)
| |
Collapse
|
6
|
Chen C, Shen J, Yang L, Zhang W, Xia R, Huan F, Gong X, Wang L, Wang C, Yuan H, Wang SL. Identification of structural properties influencing the metabolism of polycyclic aromatic hydrocarbons by cytochrome P450 1A1. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 758:143997. [PMID: 33333309 DOI: 10.1016/j.scitotenv.2020.143997] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/14/2020] [Accepted: 11/15/2020] [Indexed: 06/12/2023]
Abstract
Cytochrome P450 1A1 (CYP1A1) has served as a known metabolic enzyme that mediates the carcinogenesis of polycyclic aromatic hydrocarbons (PAHs). However, the structural mechanism involved in the metabolic capacity remains unclear. In this study, thirty-three calculated properties representing the physicochemical and electronic properties of PAH and PAH-CYP1A1 interactions were utilized to identify the key structural properties that affect metabolic processes, including binding ability, metabolic clearance, and mutagenicity, using a quantitative structure-activity relationship (QSAR) strategy combined with docking methods, QM/MM calculations and ab initio calculations. van der Waals interactions (glide vdw) appeared to be important for PAH binding to CYP1A1 and were mainly affected by the molecular weight and hydrophobic structures of PAHs. Interaction features between PAHs and heme, including the distance between iron and carbons of PAHs (Fe_Cmin) and heme vdw, coordinately influence the metabolic clearance of PAHs. Furthermore, the electronic properties (ESP neg variance) appeared to be critical for the mutagenicity of PAHs by CYP1A1 through influencing epoxide metabolite formation. The QSAR models with these key properties provide a new perspective on the structural mechanism of PAH metabolism and provide a useful in silico tool for screening, classifying and predicting PAHs for their metabolism-related toxicities and risk assessment in the environment.
Collapse
Affiliation(s)
- Chao Chen
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Jiemiao Shen
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Liu Yang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China; State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Wen Zhang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Rong Xia
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Fei Huan
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Xing Gong
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Li Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Chao Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China; State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Haoliang Yuan
- State Key Laboratory of Natural Medicines and Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shou-Lin Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China; State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China.
| |
Collapse
|
7
|
Child SA, Guengerich FP. Multistep Binding of the Non-Steroidal Inhibitors Orteronel and Seviteronel to Human Cytochrome P450 17A1 and Relevance to Inhibition of Enzyme Activity. J Med Chem 2020; 63:6513-6522. [PMID: 32223238 DOI: 10.1021/acs.jmedchem.9b01849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Orteronel (TAK-700) is a substituted imidazole that was developed for the treatment of castration-resistant prostate cancer but was dropped in phase III clinical trials. Both enantiomers of this inhibitor of cytochrome P450 (P450) 17A1 show some selectivity in differentially blocking the 17α-hydroxylation and lyase activities of the enzyme. Although both enantiomers of this compound have sub-micromolar IC50 values and bind to the enzyme with a type II spectral change (indicative of nitrogen-iron bonding) and reported Kd values of 56 and 40 nM (R and S, respectively), the rates of binding to P450 17A1 were relatively slow. We considered the possibility that the drug is a slow, tight-binding inhibitor. Analysis of the kinetics of binding revealed rapid formation of an initial complex, presumably in the substrate binding site, followed by a slower change to the spectrum of a final iron complex. Similar kinetics were observed in the interaction of another inhibitor, the triazole (S)-seviteronel (VT-464), with P450 17A1. Kinetic tests and modeling indicate that the further change to the iron-complexed form of the orteronel- or seviteronel-P450 complex is not a prerequisite for enzyme inhibition. Accordingly, the inclusion of heme-binding heterocyclic nitrogen moieties in P450 17A1 inhibitors may not be necessary to achieve inhibition but may nevertheless augment the process.
Collapse
Affiliation(s)
- Stella A Child
- Department of Biochemistry, Vanderbilt University School of Medicine, 638B Robinson Research Building, 2200 Pierce Avenue, Nashville, Tennessee 37232-0146, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, 638B Robinson Research Building, 2200 Pierce Avenue, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
8
|
Nagayoshi H, Murayama N, Tsujino M, Takenaka S, Katahira J, Kim V, Kim D, Komori M, Yamazaki H, Guengerich FP, Shimada T. Preference for O-demethylation reactions in the oxidation of 2'-, 3'-, and 4'-methoxyflavones by human cytochrome P450 enzymes. Xenobiotica 2020; 50:1158-1169. [PMID: 32312164 DOI: 10.1080/00498254.2020.1759157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
2'-, 3'-, and 4'-Methoxyflavones (MeFs) were incubated with nine forms of recombinant human cytochrome P450 (P450 or CYP) enzymes in the presence of an NADPH-generating system and the products formed were analyzed with LC-MS/MS methods.CYP1B1.1 and 1B1.3 were highly active in demethylating 4'MeF to form 4'-hydroxyflavone (rate of 5.0 nmol/min/nmol P450) and further to 3',4'-dihydroxyflavone (rates of 2.1 and 0.66 nmol/min/nmol P450, respectively). 3'MeF was found to be oxidized by P450s to m/z 239 (M-14) products (presumably 3'-hydroxyflavone) and then to 3',4'-dihydroxyflavone. P450s also catalyzed oxidation of 2'MeF to m/z 239 (M-14) and m/z 255 (M-14, M-14 + 16) products, presumably mono- and di-hydroxylated products, respectively.At least two types of ring oxidation products having m/z 269 fragments were formed, although at slower rates than the formation of mono- and di-hydroxylated products, on incubation of these MeFs with P450s; one type was products oxidized at the C-ring, having m/z 121 fragments, and the other one was the products oxidized at the A-ring (having m/z 137 fragments).Molecular docking analysis indicated the preference of interaction of O-methoxy moiety of methoxyflavones in the active site of CYP1A2.These results suggest that 2'-, 3'-, and 4'-methoxyflavones are principally demethylated by human P450s to form mono- and di-hydroxyflavones and that direct oxidation occurs in these MeFs to form mono-hydroxylated products, oxidized at the A- or B-ring of MeF.
Collapse
Affiliation(s)
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | | | - Shigeo Takenaka
- Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Habikino, Osaka, Japan
| | - Jun Katahira
- Laboratory of Cellular and Molecular Biology, Veterinary Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Vitchan Kim
- Department of Biological Sciences, Konkuk University, Seoul, Korea, and
| | - Donghak Kim
- Department of Biological Sciences, Konkuk University, Seoul, Korea, and
| | - Masayuki Komori
- Laboratory of Cellular and Molecular Biology, Veterinary Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Veterinary Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan
| |
Collapse
|
9
|
Bart AG, Harris KL, Gillam EMJ, Scott EE. Structure of an ancestral mammalian family 1B1 cytochrome P450 with increased thermostability. J Biol Chem 2020; 295:5640-5653. [PMID: 32156703 PMCID: PMC7186169 DOI: 10.1074/jbc.ra119.010727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/09/2020] [Indexed: 01/07/2023] Open
Abstract
Mammalian cytochrome P450 enzymes often metabolize many pharmaceuticals and other xenobiotics, a feature that is valuable in a biotechnology setting. However, extant P450 enzymes are typically relatively unstable, with T50 values of ∼30-40 °C. Reconstructed ancestral cytochrome P450 enzymes tend to have variable substrate selectivity compared with related extant forms, but they also have higher thermostability and therefore may be excellent tools for commercial biosynthesis of important intermediates, final drug molecules, or drug metabolites. The mammalian ancestor of the cytochrome P450 1B subfamily was herein characterized structurally and functionally, revealing differences from the extant human CYP1B1 in ligand binding, metabolism, and potential molecular contributors to its thermostability. Whereas extant human CYP1B1 has one molecule of α-naphthoflavone in a closed active site, we observed that subtle amino acid substitutions outside the active site in the ancestor CYP1B enzyme yielded an open active site with four ligand copies. A structure of the ancestor with 17β-estradiol revealed only one molecule in the active site, which still had the same open conformation. Detailed comparisons between the extant and ancestor forms revealed increases in electrostatic and aromatic interactions between distinct secondary structure elements in the ancestral forms that may contribute to their thermostability. To the best of our knowledge, this represents the first structural evaluation of a reconstructed ancestral cytochrome P450, revealing key features that appear to contribute to its thermostability.
Collapse
Affiliation(s)
- Aaron G Bart
- Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109
| | - Kurt L Harris
- School of Chemistry and Molecular Biosciences, University of Queensland St. Lucia, Brisbane 4072, Australia
| | - Elizabeth M J Gillam
- School of Chemistry and Molecular Biosciences, University of Queensland St. Lucia, Brisbane 4072, Australia
| | - Emily E Scott
- Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109; Departments of Medicinal Chemistry and Pharmacology, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
10
|
Cervellati F, Benedusi M, Manarini F, Woodby B, Russo M, Valacchi G, Pietrogrande MC. Proinflammatory properties and oxidative effects of atmospheric particle components in human keratinocytes. CHEMOSPHERE 2020; 240:124746. [PMID: 31568946 DOI: 10.1016/j.chemosphere.2019.124746] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 06/10/2023]
Abstract
The skin is one of the main organs exposed to airborne particulate matter (PM), which may contain various pollutants linked to a wide range of adverse health endpoints. In the present work, we analyzed the proinflammatory and oxidative effects of some PM components leading to inflammatory responses, cell proliferation or cell death. We investigated four redox-active chemicals, such as Cu (II) metal and quinones generated from polycyclic aromatic hydrocarbons (PAHs), i.e., 9,10 phenanthrenequinone and isomers 1,2 and 1,4 naphthoquinone. We performed in vitro biological tests on human keratinocyte (HaCaT) cells and also acellular assays based on the oxidation of dithiothreitol and ascorbic acid, antioxidants to assess the oxidative potential (OP). We found that treated keratinocytes showed increased activation of the redox-sensitive transcription factor NFκB and increased transcript levels of the NFκB-dependent gene IL8. Moreover, the treatment with Cu(II) and quinones increased the activities and the expression of genes involved in the redox response, SOD1 and GPX, suggesting that PM components induced cellular damage due to redox imbalances. Finally, we found alteration of the mitochondrial ultrastructure and increased apoptosis after 24 h of treatment. The results presented suggest that all of the analyzed pollutant components are able to modulate similar signal transduction pathways, resulting in activation of inflammatory processes in the skin, followed by oxidative damage. Altogether these observations indicate that exposure of skin to air pollutants modifies the redox equilibrium of keratinocytes, which could explain the increased skin damage observed in populations that live in high-pollution cities.
Collapse
Affiliation(s)
- Franco Cervellati
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, NC State University, Siena, Italy.
| | - Mascia Benedusi
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, NC State University, Siena, Italy
| | - Francesco Manarini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Brittany Woodby
- Plants for Human Health Institute Animal Science Dept., NC Research Campus, Kannapolis, NC, 28081, USA
| | - Mara Russo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, NC State University, Siena, Italy; Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute Animal Science Dept., NC Research Campus, Kannapolis, NC, 28081, USA; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | | |
Collapse
|
11
|
Bao L, Liu W, Li Y, Wang X, Xu F, Yang Z, Yue Y, Zuo C, Zhang Q, Wang W. Carcinogenic Metabolic Activation Process of Naphthalene by the Cytochrome P450 Enzyme 1B1: A Computational Study. Chem Res Toxicol 2019; 32:603-612. [PMID: 30794404 DOI: 10.1021/acs.chemrestox.8b00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The metabolic activation and transformation of naphthalene by the cytochrome P450 enzyme (CYP 1B1) plays an important role in its potential carcinogenicity. The process has been explored by a quantum mechanics/molecular mechanics (QM/MM) computational method. Molecular dynamic simulations were performed to explore the interaction between naphthalene and CYP 1B1. Naphthalene involves α- and β-carbon, the electrophilic addition of which would result in different reaction pathways. Our computational results show that both additions on α- and β-carbon can generate naphthalene 1,2-oxide. The activation barrier for the addition on β-carbon is higher than that for the α-carbon by 2.6 kcal·mol-1, which is possibly caused by the proximity between β-carbon and the iron-oxo group of Cpd I in the system. We also found that naphthalene 1,2-oxide is unstable and the O-C bond cleavage easily occurs via cellular hydronium ion, hydroxyl radical/anion; then it will convert to the potential ultimate carcinogen 1,2-naphthoquinone. The results demonstrate and inform a detailed process of generating naphthalene 1,2-oxide and new predictions for its conversion.
Collapse
Affiliation(s)
- Lei Bao
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Wen Liu
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Yanwei Li
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Xueyu Wang
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Fei Xu
- Shenzhen Research Institute of Shandong University , Shenzhen 518057 , People's Republic of China
| | - Zhongyue Yang
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Yue Yue
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Chenpeng Zuo
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Qingzhu Zhang
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| | - Wenxing Wang
- Environment Research Institute , Shandong University , Qingdao 266237 , People's Republic of China
| |
Collapse
|
12
|
Mohd Siddique MU, Barbhuiya TK, Sinha BN, Jayaprakash V. Phytoestrogens and their synthetic analogues as substrate mimic inhibitors of CYP1B1. Eur J Med Chem 2019; 163:28-36. [DOI: 10.1016/j.ejmech.2018.11.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
|
13
|
Nagayoshi H, Murayama N, Kakimoto K, Takenaka S, Katahira J, Lim YR, Kim V, Kim D, Yamazaki H, Komori M, Guengerich FP, Shimada T. Site-specific oxidation of flavanone and flavone by cytochrome P450 2A6 in human liver microsomes. Xenobiotica 2018; 49:791-802. [PMID: 30048196 DOI: 10.1080/00498254.2018.1505064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The roles of human cytochrome P450 (P450 or CYP) 2A6 in the oxidation of flavanone [(2R)- and (2S)-enantiomers] and flavone were studied in human liver microsomes and recombinant human P450 enzymes. CYP2A6 was highly active in oxidizing flavanone to form flavone, 2'-hydroxy-, 4'-, and 6-hydroxyflavanones and in oxidizing flavone to form mono- and di-hydroxylated products, such as mono-hydroxy flavones M6, M7, and M11 and di-hydroxy flavones M3, M4, and M5. Liver microsomes prepared from human sample HH2, defective in coumarin 7-hydroxylation activity, were very inefficient in forming 2'-hydroxyflavanone from flavanone and a mono-hydroxylated product, M6, from flavone. Coumarin and anti-CYP2A6 antibodies strongly inhibited the formation of these metabolites in microsomes prepared from liver samples HH47 and 54, which were active in coumarin oxidation activities. Molecular docking analysis showed that the C2'-position of (2R)-flavanone (3.8 Å) was closer to the iron center of CYP2A6 than the C6-position (10 Å), while distances from C2' and C6 of (2S)-flavanone to the CYP2A6 were 6.91 Å and 5.42 Å, respectively. These results suggest that CYP2A6 catalyzes site-specific oxidation of (racemic) flavanone and also flavone in human liver microsomes. CYP1A2 and CYP2B6 were also found to play significant roles in some of the oxidations of these flavonoids by human liver microsomes.
Collapse
Affiliation(s)
| | - Norie Murayama
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo , Japan
| | | | - Shigeo Takenaka
- c Graduate School of Comprehensive Rehabilitation , Osaka Prefecture University , Habikino Osaka , Japan
| | - Jun Katahira
- d Laboratory of Cellular and Molecular Biology , Veterinary Sciences, Osaka Prefecture University , Izumisano , Osaka , Japan
| | - Young-Ran Lim
- e Department of Biological Sciences , Konkuk University , Seoul , Korea
| | - Vitchan Kim
- e Department of Biological Sciences , Konkuk University , Seoul , Korea
| | - Donghak Kim
- e Department of Biological Sciences , Konkuk University , Seoul , Korea
| | - Hiroshi Yamazaki
- b Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo , Japan
| | - Masayuki Komori
- d Laboratory of Cellular and Molecular Biology , Veterinary Sciences, Osaka Prefecture University , Izumisano , Osaka , Japan
| | - F Peter Guengerich
- f Department of Biochemistry Vanderbilt University School of Medicine , Nashville , Tennessee , USA
| | - Tsutomu Shimada
- d Laboratory of Cellular and Molecular Biology , Veterinary Sciences, Osaka Prefecture University , Izumisano , Osaka , Japan
| |
Collapse
|
14
|
Jeffreys LN, Girvan HM, McLean KJ, Munro AW. Characterization of Cytochrome P450 Enzymes and Their Applications in Synthetic Biology. Methods Enzymol 2018; 608:189-261. [PMID: 30173763 DOI: 10.1016/bs.mie.2018.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cytochrome P450 monooxygenase enzymes (P450s) catalyze a diverse array of chemical transformations, most originating from the insertion of an oxygen atom into a substrate that binds close to the P450 heme. The oxygen is delivered by a highly reactive heme iron-oxo species (compound I) and, according to the chemical nature of the substrate and its position in the active site, the P450 can catalyze a wide range of reactions including, e.g., hydroxylation, reduction, decarboxylation, sulfoxidation, N- and O-demethylation, epoxidation, deamination, CC bond formation and breakage, nitration, and dehalogenation. In this chapter, we describe the structural, biochemical, and catalytic properties of the P450s, along with spectroscopic and analytical methods used to characterize P450 enzymes and their redox partners. Important uses of P450 enzymes are highlighted, including how various P450s have been exploited for applications in synthetic biology.
Collapse
Affiliation(s)
- Laura N Jeffreys
- Manchester Institute of Biotechnology, University of Manchester, Manchester, United Kingdom
| | - Hazel M Girvan
- Manchester Institute of Biotechnology, University of Manchester, Manchester, United Kingdom
| | - Kirsty J McLean
- Manchester Institute of Biotechnology, University of Manchester, Manchester, United Kingdom
| | - Andrew W Munro
- Manchester Institute of Biotechnology, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
15
|
Lockart MM, Rodriguez CA, Atkins WM, Bowman MK. CW EPR parameters reveal cytochrome P450 ligand binding modes. J Inorg Biochem 2018. [PMID: 29530595 DOI: 10.1016/j.jinorgbio.2018.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cytochrome P450 (CYP) monoxygenses utilize heme cofactors to catalyze oxidation reactions. They play a critical role in metabolism of many classes of drugs, are an attractive target for drug development, and mediate several prominent drug interactions. Many substrates and inhibitors alter the spin state of the ferric heme by displacing the heme's axial water ligand in the resting enzyme to yield a five-coordinate iron complex, or they replace the axial water to yield a nitrogen-ligated six-coordinate iron complex, which are traditionally assigned by UV-vis spectroscopy. However, crystal structures and recent pulsed electron paramagnetic resonance (EPR) studies find a few cases where molecules hydrogen bond to the axial water. The water-bridged drug-H2O-heme has UV-vis spectra similar to nitrogen-ligated, six-coordinate complexes, but are closer to "reverse type I" complexes described in older liteature. Here, pulsed and continuous wave (CW) EPR demonstrate that water-bridged complexes are remarkably common among a range of nitrogenous drugs or drug fragments that bind to CYP3A4 or CYP2C9. Principal component analysis reveals a distinct clustering of CW EPR spectral parameters for water-bridged complexes. CW EPR reveals heterogeneous mixtures of ligated states, including multiple directly-coordinated complexes and water-bridged complexes. These results suggest that water-bridged complexes are under-represented in CYP structural databases and can have energies similar to other ligation modes. The data indicates that water-bridged binding modes can be identified and distinguished from directly-coordinated binding by CW EPR.
Collapse
Affiliation(s)
- Molly M Lockart
- Department of Chemistry and Biochemistry, Box 870336, University of Alabama, Tuscaloosa, AL 35487-0336, United States
| | - Carlo A Rodriguez
- Department of Chemistry and Biochemistry, Box 870336, University of Alabama, Tuscaloosa, AL 35487-0336, United States
| | - William M Atkins
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, WA 98195-7610, United States
| | - Michael K Bowman
- Department of Chemistry and Biochemistry, Box 870336, University of Alabama, Tuscaloosa, AL 35487-0336, United States.
| |
Collapse
|
16
|
Traylor MJ, Baek JM, Richards KE, Fusetto R, Huang W, Josh P, Chen Z, Bollapragada P, O'Hair RAJ, Batterham P, Gillam EMJ. Recombinant expression and characterization of Lucilia cuprina CYP6G3: Activity and binding properties toward multiple pesticides. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2017; 90:14-22. [PMID: 28918158 DOI: 10.1016/j.ibmb.2017.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 06/07/2023]
Abstract
The Australian sheep blowfly, Lucilia cuprina, is a primary cause of sheep flystrike and a major agricultural pest. Cytochrome P450 enzymes have been implicated in the resistance of L. cuprina to several classes of insecticides. In particular, CYP6G3 is a L. cuprina homologue of Drosophila melanogaster CYP6G1, a P450 known to confer multi-pesticide resistance. To investigate the basis of resistance, a bicistronic Escherichia coli expression system was developed to co-express active L. cuprina CYP6G3 and house fly (Musca domestica) P450 reductase. Recombinant CYP6G3 showed activity towards the high-throughput screening substrates, 7-ethoxycoumarin and p-nitroanisole, but not towards p-nitrophenol, coumarin, 7-benzyloxyresorufin, or seven different luciferin derivatives (P450-Glo™ substrates). The addition of house fly cytochrome b5 enhanced the kcat for p-nitroanisole dealkylation approximately two fold (17.8 ± 0.5 vs 9.6 ± 0.2 min-1) with little effect on KM (13 ± 1 vs 10 ± 1 μM). Inhibition studies and difference spectroscopy revealed that the organochlorine compounds, DDT and endosulfan, and the organophosphate pesticides, malathion and chlorfenvinphos, bind to the active site of CYP6G3. All four pesticides showed type I binding spectra with spectral dissociation constants in the micromolar range suggesting that they may be substrates of CYP6G3. While no significant inhibition was seen with the organophosphate, diazinon, or the neonicotinoid, imidacloprid, diazinon showed weak binding in spectral assays, with a Kd value of 23 ± 3 μM CYP6G3 metabolised diazinon to the diazoxon and hydroxydiazinon metabolites and imidacloprid to the 5-hydroxy and olefin metabolites, consistent with a proposed role of CYP6G enzymes in metabolism of phosphorothioate and neonicotinoid insecticides in other species.
Collapse
Affiliation(s)
- Matthew J Traylor
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Jong-Min Baek
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Katelyn E Richards
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Roberto Fusetto
- The Bio21 Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - W Huang
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Peter Josh
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Zhenzhong Chen
- The Bio21 Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Padma Bollapragada
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia
| | - Richard A J O'Hair
- The Bio21 Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Philip Batterham
- The Bio21 Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Elizabeth M J Gillam
- School of Chemistry and Molecular Biology, University of Queensland, St. Lucia 4072, Australia.
| |
Collapse
|
17
|
Shimada T, Murayama N, Kakimoto K, Takenaka S, Lim YR, Yeom S, Kim D, Yamazaki H, Guengerich FP, Komori M. Oxidation of 1-chloropyrene by human CYP1 family and CYP2A subfamily cytochrome P450 enzymes: catalytic roles of two CYP1B1 and five CYP2A13 allelic variants. Xenobiotica 2017. [PMID: 28648140 DOI: 10.1080/00498254.2017.1347306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
1. 1-Chloropyrene, one of the major chlorinated polycyclic aromatic hydrocarbon contaminants, was incubated with human cytochrome P450 (P450 or CYP) enzymes including CYP1A1, 1A2, 1B1, 2A6, 2A13, 2B6, 2C9, 2D6, 2E1, 3A4 and 3A5. Catalytic differences in 1-chloropyrene oxidation by polymorphic two CYP1B1 and five CYP2A13 allelic variants were also examined. 2. CYP1A1 oxidized 1-chloropyrene at the 6- and 8-positions more actively than at the 3-position, while both CYP1B1.1 and 1B1.3 preferentially catalyzed 6-hydroxylation. 3. Five CYP2A13 allelic variants oxidized 8-hydroxylation much more than 6- and 3-hydroxylation, and the variant CYP2A13.3 was found to slowly catalyze these reactions with a lower kcat value than other CYP2A13.1 variants. 4. CYP2A6 catalyzed 1-chloropyrene 6-hydroxylation at a higher rate than the CYP2A13 enzymes, but the rate was lower than the CYP1A1 and 1B1 variants. Other human P450 enzymes had low activities towards 1-chloropyrene. 5. Molecular docking analysis suggested differences in the interaction of 1-chloropyrene with active sites of CYP1 and 2 A enzymes. In addition, a naturally occurring Thr134 insertion in CYP2A13.3 was found to affect the orientation of Asn297 in the I-helix in interacting with 1-chloropyrene (and also 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, NNK) and caused changes in the active site of CYP2A13.3 as compared with CYP2A13.1.
Collapse
Affiliation(s)
- Tsutomu Shimada
- a Laboratory of Cellular and Molecular Biology, Osaka Prefecture University , Osaka , Japan
| | - Norie Murayama
- b Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Machida , Tokyo
| | | | - Shigeo Takenaka
- d Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University , Osaka , Japan
| | - Young-Ran Lim
- e Department of Biological Sciences , Konkuk University , Seoul , Korea , and
| | - Sora Yeom
- e Department of Biological Sciences , Konkuk University , Seoul , Korea , and
| | - Donghak Kim
- e Department of Biological Sciences , Konkuk University , Seoul , Korea , and
| | - Hiroshi Yamazaki
- b Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University , Machida , Tokyo
| | - F Peter Guengerich
- f Department of Biochemistry , Vanderbilt University School of Medicine , Nashville, TN , USA
| | - Masayuki Komori
- a Laboratory of Cellular and Molecular Biology, Osaka Prefecture University , Osaka , Japan
| |
Collapse
|
18
|
Dutour R, Poirier D. Inhibitors of cytochrome P450 (CYP) 1B1. Eur J Med Chem 2017; 135:296-306. [DOI: 10.1016/j.ejmech.2017.04.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/13/2017] [Accepted: 04/17/2017] [Indexed: 12/16/2022]
|
19
|
Shimada T. Inhibition of Carcinogen-Activating Cytochrome P450 Enzymes by Xenobiotic Chemicals in Relation to Antimutagenicity and Anticarcinogenicity. Toxicol Res 2017; 33:79-96. [PMID: 28443179 PMCID: PMC5402866 DOI: 10.5487/tr.2017.33.2.079] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/16/2017] [Indexed: 12/27/2022] Open
Abstract
A variety of xenobiotic chemicals, such as polycyclic aromatic hydrocarbons (PAHs), aryl- and heterocyclic amines and tobacco related nitrosamines, are ubiquitous environmental carcinogens and are required to be activated to chemically reactive metabolites by xenobiotic-metabolizing enzymes, including cytochrome P450 (P450 or CYP), in order to initiate cell transformation. Of various human P450 enzymes determined to date, CYP1A1, 1A2, 1B1, 2A13, 2A6, 2E1, and 3A4 are reported to play critical roles in the bioactivation of these carcinogenic chemicals. In vivo studies have shown that disruption of Cyp1b1 and Cyp2a5 genes in mice resulted in suppression of tumor formation caused by 7,12-dimethylbenz[a]anthracene and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, respectively. In addition, specific inhibitors for CYP1 and 2A enzymes are able to suppress tumor formation caused by several carcinogens in experimental animals in vivo, when these inhibitors are applied before or just after the administration of carcinogens. In this review, we describe recent progress, including our own studies done during past decade, on the nature of inhibitors of human CYP1 and CYP2A enzymes that have been shown to activate carcinogenic PAHs and tobacco-related nitrosamines, respectively, in humans. The inhibitors considered here include a variety of carcinogenic and/or non-carcinogenic PAHs and acethylenic PAHs, many flavonoid derivatives, derivatives of naphthalene, phenanthrene, biphenyl, and pyrene and chemopreventive organoselenium compounds, such as benzyl selenocyanate and benzyl selenocyanate; o-XSC, 1,2-, 1,3-, and 1,4-phenylenebis( methylene)selenocyanate.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Veterinary Sciences, Osaka Prefecture University, Osaka, Japan
| |
Collapse
|
20
|
Shimada T, Kakimoto K, Takenaka S, Koga N, Uehara S, Murayama N, Yamazaki H, Kim D, Guengerich FP, Komori M. Roles of Human CYP2A6 and Monkey CYP2A24 and 2A26 Cytochrome P450 Enzymes in the Oxidation of 2,5,2',5'-Tetrachlorobiphenyl. Drug Metab Dispos 2016; 44:1899-1909. [PMID: 27625140 PMCID: PMC6047209 DOI: 10.1124/dmd.116.072991] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/12/2016] [Indexed: 11/22/2022] Open
Abstract
2,5,2',5'-Tetrachlorobiphenyl (TCB) induced type I binding spectra with cytochrome P450 (P450) 2A6 and 2A13, with Ks values of 9.4 and 0.51 µM, respectively. However, CYP2A6 oxidized 2,5,2',5'-TCB to form 4-hydroxylated products at a much higher rate (∼1.0 minute-1) than CYP2A13 (∼0.02 minute-1) based on analysis by liquid chromatography-tandem mass spectrometry. Formation of 4-hydroxy-2,5,2',5'-TCB by CYP2A6 was greater than that of 3-hydroxy-2,5,2',5'-TCB and three other hydroxylated products. Several human P450 enzymes, including CYP1A1, 1A2, 1B1, 2B6, 2D6, 2E1, 2C9, and 3A4, did not show any detectable activities in oxidizing 2,5,2',5'-TCB. Cynomolgus monkey CYP2A24, which shows 95% amino acid identity to human CYP2A6, catalyzed 4-hydroxylation of 2,5,2',5'-TCB at a higher rate (∼0.3 minute-1) than CYP2A26 (93% identity to CYP2A6, ∼0.13 minute-1) and CYP2A23 (94% identity to CYP2A13, ∼0.008 minute-1). None of these human and monkey CYP2A enzymes were catalytically active in oxidizing other TCB congeners, such as 2,4,3',4'-, 3,4,3',4'-, and 3,5,3',5'-TCB. Molecular docking analysis suggested that there are different orientations of interaction of 2,5,2',5'-TCB with the active sites (over the heme) of human and monkey CYP2A enzymes, and that ligand interaction energies (U values) of bound protein-ligand complexes show structural relationships of interaction of TCBs and other ligands with active sites of CYP2A enzymes. Catalytic differences in human and monkey CYP2A enzymes in the oxidation of 2,5,2',5'-TCB are suggested to be due to amino acid changes at substrate recognition sites, i.e., V110L, I209S, I300F, V365M, S369G, and R372H, based on the comparison of primary sequences.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Kensaku Kakimoto
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Shigeo Takenaka
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Nobuyuki Koga
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Shotaro Uehara
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Norie Murayama
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Hiroshi Yamazaki
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Donghak Kim
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - F Peter Guengerich
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| | - Masayuki Komori
- Laboratory of Cellular and Molecular Biology, Osaka Prefecture University, Izumisano, Osaka, Japan (T.S., S.T., M.K.); Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, Japan (K.K.); Faculty of Nutritional Sciences, Nakamura Gakuen University, Johnan-ku, Fukuoka, Japan (N.K.); Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., N.M., H.Y.); Department of Biological Sciences, Konkuk University, Seoul, South Korea (D.K.); and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee (F.P.G.)
| |
Collapse
|
21
|
Kumar R, Gupta D. Identification of CYP1B1-specific candidate inhibitors using combination ofin silicoscreening, integrated knowledge-based filtering, and molecular dynamics simulations. Chem Biol Drug Des 2016; 88:730-739. [DOI: 10.1111/cbdd.12803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Rakesh Kumar
- Translational Bioinformatics Group; International Centre for Genetic Engineering and Biotechnology (ICGEB); New Delhi Delhi India
| | - Dinesh Gupta
- Translational Bioinformatics Group; International Centre for Genetic Engineering and Biotechnology (ICGEB); New Delhi Delhi India
| |
Collapse
|
22
|
Shimada T, Takenaka S, Kakimoto K, Murayama N, Lim YR, Kim D, Foroozesh MK, Yamazaki H, Guengerich FP, Komori M. Structure-Function Studies of Naphthalene, Phenanthrene, Biphenyl, and Their Derivatives in Interaction with and Oxidation by Cytochromes P450 2A13 and 2A6. Chem Res Toxicol 2016; 29:1029-40. [PMID: 27137136 PMCID: PMC5293596 DOI: 10.1021/acs.chemrestox.6b00083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Naphthalene, phenanthrene, biphenyl, and their derivatives having different ethynyl, propynyl, butynyl, and propargyl ether substitutions were examined for their interaction with and oxidation by cytochromes P450 (P450) 2A13 and 2A6. Spectral interaction studies suggested that most of these chemicals interacted with P450 2A13 to induce Type I binding spectra more readily than with P450 2A6. Among the various substituted derivatives examined, 2-ethynylnaphthalene, 2-naphthalene propargyl ether, 3-ethynylphenanthrene, and 4-biphenyl propargyl ether had larger ΔAmax/Ks values in inducing Type I binding spectra with P450 2A13 than their parent compounds. P450 2A13 was found to oxidize naphthalene, phenanthrene, and biphenyl to 1-naphthol, 9-hydroxyphenanthrene, and 2- and/or 4-hydroxybiphenyl, respectively, at much higher rates than P450 2A6. Other human P450 enzymes including P450s 1A1, 1A2, 1B1, 2C9, and 3A4 had lower rates of oxidation of naphthalene, phenanthrene, and biphenyl than P450s 2A13 and 2A6. Those alkynylated derivatives that strongly induced Type I binding spectra with P450s 2A13 and 2A6 were extensively oxidized by these enzymes upon analysis with HPLC. Molecular docking studies supported the hypothesis that ligand-interaction energies (U values) obtained with reported crystal structures of P450 2A13 and 2A6 bound to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, indole, pilocarpine, nicotine, and coumarin are of use in understanding the basis of possible molecular interactions of these xenobiotic chemicals with the active sites of P450 2A13 and 2A6 enzymes. In fact, the ligand-interaction energies with P450 2A13 4EJG bound to these chemicals were found to relate to their induction of Type I binding spectra.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Shigeo Takenaka
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Kensaku Kakimoto
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Young-Ran Lim
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Donghak Kim
- Department of Biological Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Maryam K. Foroozesh
- Department of Chemistry, Xavier University of Louisiana, New Orleans, Louisiana 70125, United States
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Masayuki Komori
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
23
|
Korobkova EA. Effect of Natural Polyphenols on CYP Metabolism: Implications for Diseases. Chem Res Toxicol 2015; 28:1359-90. [PMID: 26042469 DOI: 10.1021/acs.chemrestox.5b00121] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cytochromes P450 (CYPs) are a large group of hemeproteins located on mitochondrial membranes or the endoplasmic reticulum. They play a crucial role in the metabolism of endogenous and exogenous molecules. The activity of CYP is associated with a number of factors including redox potential, protein conformation, the accessibility of the active site by substrates, and others. This activity may be potentially modulated by a variety of small molecules. Extensive experimental data collected over the past decade point at the active role of natural polyphenols in modulating the catalytic activity of CYP. Polyphenols are widespread micronutrients present in human diets of plant origin and in medicinal herbs. These compounds may alter the activity of CYP either via direct interactions with the enzymes or by affecting CYP gene expression. The polyphenol-CYP interactions may significantly alter the pharmacokinetics of drugs and thus influence the effectiveness of chemical therapies used in the treatment of different types of cancers, diabetes, obesity, and cardiovascular diseases (CVD). CYPs are involved in the oxidation and activation of external carcinogenic agents, in which case the inhibition of the CYP activity is beneficial for health. CYPs also support detoxification processes. In this case, it is the upregulation of CYP genes that would be favorable for the organism. A CYP enzyme aromatase catalyzes the formation of estrone and estradiol from their precursors. CYPs also catalyze multiple reactions leading to the oxidation of estrogen. Estrogen signaling and oxidative metabolism of estrogen are associated with the development of cancer. Thus, polyphenol-mediated modulation of the CYP's activity also plays a vital role in estrogen carcinogenesis. The aim of the present review is to summarize the data collected over the last five to six years on the following topics: (1) the mechanisms of the interactions of CYP with food constituents that occur via the direct binding of polyphenols to the enzymes and (2) the mechanisms of the regulation of CYP gene expression mediated by polyphenols. The structure-activity relationship relevant to the ability of polyphenols to affect the activity of CYP is analyzed. The application of polyphenol-CYP interactions to diseases is discussed.
Collapse
Affiliation(s)
- Ekaterina A Korobkova
- John Jay College of Criminal Justice, The Department of Sciences, City University of New York, 524 W 59th Street, New York, New York 10019, United States
| |
Collapse
|
24
|
Conner KP, Cruce AA, Krzyaniak MD, Schimpf AM, Frank DJ, Ortiz de Montellano P, Atkins WM, Bowman MK. Drug modulation of water-heme interactions in low-spin P450 complexes of CYP2C9d and CYP125A1. Biochemistry 2015; 54:1198-207. [PMID: 25591012 DOI: 10.1021/bi501402k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Azoles and pyridines are commonly incorporated into small molecule inhibitor scaffolds that target cytochromes P450 (CYPs) as a strategy to increase drug binding affinity, impart isoform-dependent selectivity, and improve metabolic stability. Optical absorbance spectra of the CYP-inhibitor complex are widely used to infer whether these inhibitors are ligated directly to the heme iron as catalytically inert, low-spin (type II) complexes. Here, we show that the low-spin complex between a drug-metabolizing CYP2C9 variant and 4-(3-phenylpropyl)-1H-1,2,3-triazole (PPT) retains an axial water ligand despite exhibiting elements of "classic" type II optical behavior. Hydrogens of the axial water ligand are observed by pulsed electron paramagnetic resonance (EPR) spectroscopy for both inhibitor-free and inhibitor-bound species and show that inhibitor binding does not displace the axial water. A (15)N label incorporated into PPT is 0.444 nm from the heme iron, showing that PPT is also in the active site. The reverse type I inhibitor, LP10, of CYP125A1 from Mycobacterium tuberculosis, known from X-ray crystal structures to form a low-spin water-bridged complex, is found by EPR and by visible and near-infrared magnetic circular dichroism spectroscopy to retain the axial water ligand in the complex in solution.
Collapse
Affiliation(s)
- Kip P Conner
- Department of Medicinal Chemistry, Box 357610, and Department of Chemistry, Box 351700, University of Washington , Seattle, Washington 98195, United States
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim D, Cha GS, Nagy LD, Yun CH, Guengerich FP. Kinetic analysis of lauric acid hydroxylation by human cytochrome P450 4A11. Biochemistry 2014; 53:6161-72. [PMID: 25203493 PMCID: PMC4188250 DOI: 10.1021/bi500710e] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Cytochrome P450 (P450) 4A11 is the
only functionally active subfamily
4A P450 in humans. P450 4A11 catalyzes mainly ω-hydroxylation
of fatty acids in liver and kidney; this process is not a major degradative
pathway, but at least one product, 20-hydroxyeicosatetraenoic acid,
has important signaling properties. We studied catalysis by P450 4A11
and the issue of rate-limiting steps using lauric acid ω-hydroxylation,
a prototypic substrate for this enzyme. Some individual reaction steps
were studied using pre-steady-state kinetic approaches. Substrate
and product binding and release were much faster than overall rates
of catalysis. Reduction of ferric P450 4A11 (to ferrous) was rapid
and not rate-limiting. Deuterium kinetic isotope effect (KIE) experiments
yielded low but reproducible values (1.2–2) for 12-hydroxylation
with 12-2H-substituted lauric acid. However, considerable
“metabolic switching” to 11-hydroxylation was observed
with [12-2H3]lauric acid. Analysis of switching
results [Jones, J. P., et al. (1986) J. Am. Chem. Soc.108, 7074–7078] and the use of tritium KIE
analysis with [12-3H]lauric acid [Northrop, D. B. (1987) Methods Enzymol.87, 607–625] both
indicated a high intrinsic KIE (>10). Cytochrome b5 (b5) stimulated steady-state
lauric acid ω-hydroxylation ∼2-fold; the apoprotein was
ineffective, indicating that electron transfer is involved in the b5 enhancement. The rate of b5 reoxidation was increased in the presence of ferrous
P450 mixed with O2. Collectively, the results indicate
that both the transfer of an electron to the ferrous·O2 complex and C–H bond-breaking limit the rate of P450 4A11
ω-oxidation.
Collapse
Affiliation(s)
- Donghak Kim
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | |
Collapse
|
26
|
Cytochrome P450 family 1 inhibitors and structure-activity relationships. Molecules 2013; 18:14470-95. [PMID: 24287985 PMCID: PMC4216474 DOI: 10.3390/molecules181214470] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/15/2013] [Accepted: 11/19/2013] [Indexed: 12/28/2022] Open
Abstract
With the widespread use of O-alkoxyresorufin dealkylation assays since the 1990s, thousands of inhibitors of cytochrome P450 family 1 enzymes (P450s 1A1, 1A2, and 1B1) have been identified and studied. Generally, planar polycyclic molecules such as polycyclic aromatic hydrocarbons, stilbenoids, and flavonoids are considered to potentially be effective inhibitors of these enzymes, however, the details of the structure-activity relationships and selectivity of these inhibitors are still ambiguous. In this review, we thoroughly discuss the selectivity of many representative P450 family 1 inhibitors reported in the past 20 years through a meta-analysis.
Collapse
|
27
|
Shimada T, Murayama N, Yamazaki H, Tanaka K, Takenaka S, Komori M, Kim D, Guengerich FP. Metabolic activation of polycyclic aromatic hydrocarbons and aryl and heterocyclic amines by human cytochromes P450 2A13 and 2A6. Chem Res Toxicol 2013; 26:529-37. [PMID: 23432465 DOI: 10.1021/tx3004906] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. P450 2A6 also interacted with acenaphthene and acenaphthylene, but not with fluoranthene, fluoranthene-2,3-diol, or 1-nitropyrene. P450 1B1 is well-known to oxidize many carcinogenic PAHs, and we found that several PAHs (i.e., 7,12-dimethylbenz[a]anthracene, 7,12-dimethylbenz[a]anthracene-5,6-diol, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, 5-methylchrysene, benz[a]pyrene-4,5-diol, benzo[a]pyrene-7,8-diol, 1-nitropyrene, 2-aminoanthracene, 2-aminofluorene, and 2-acetylaminofluorene) interacted with P450 1B1, producing Reverse Type I binding spectra. Metabolic activation of PAHs and aryl- and heterocyclic amines to genotoxic products was examined in Salmonella typhimurium NM2009, and we found that P450 2A13 and 2A6 (as well as P450 1B1) were able to activate several of these procarcinogens. The former two enzymes were particularly active in catalyzing 2-aminofluorene and 2-aminoanthracene activation, and molecular docking simulations supported the results with these procarcinogens, in terms of binding in the active sites of P450 2A13 and 2A6. These results suggest that P450 2A enzymes, as well as P450 Family 1 enzymes including P450 1B1, are major enzymes involved in activating PAHs and aryl- and heterocyclic amines, as well as tobacco-related nitrosamines.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Shimada T, Kim D, Murayama N, Tanaka K, Takenaka S, Nagy LD, Folkman LM, Foroozesh MK, Komori M, Yamazaki H, Guengerich FP. Binding of diverse environmental chemicals with human cytochromes P450 2A13, 2A6, and 1B1 and enzyme inhibition. Chem Res Toxicol 2013; 26:517-28. [PMID: 23432429 DOI: 10.1021/tx300492j] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A total of 68 chemicals including derivatives of naphthalene, phenanthrene, fluoranthene, pyrene, biphenyl, and flavone were examined for their abilities to interact with human P450s 2A13 and 2A6. Fifty-one of these 68 chemicals induced stronger Type I binding spectra (iron low- to high-spin state shift) with P450 2A13 than those seen with P450 2A6, i.e., the spectral binding intensities (ΔAmax/Ks ratio) determined with these chemicals were always higher for P450 2A13. In addition, benzo[c]phenanthrene, fluoranthene, 2,3-dihydroxy-2,3-dihydrofluoranthene, pyrene, 1-hydroxypyrene, 1-nitropyrene, 1-acetylpyrene, 2-acetylpyrene, 2,5,2',5'-tetrachlorobiphenyl, 7-hydroxyflavone, chrysin, and galangin were found to induce a Type I spectral change only with P450 2A13. Coumarin 7-hydroxylation, catalyzed by P450 2A13, was strongly inhibited by 2'-methoxy-5,7-dihydroxyflavone, 2-ethynylnaphthalene, 2'-methoxyflavone, 2-naphththalene propargyl ether, acenaphthene, acenaphthylene, naphthalene, 1-acetylpyrene, flavanone, chrysin, 3-ethynylphenanthrene, flavone, and 7-hydroxyflavone; these chemicals induced Type I spectral changes with low Ks values. On the basis of the intensities of the spectral changes and inhibition of P450 2A13, we classified the 68 chemicals into eight groups based on the order of affinities for these chemicals and inhibition of P450 2A13. The metabolism of chemicals by P450 2A13 during the assays explained why some of the chemicals that bound well were poor inhibitors of P450 2A13. Finally, we compared the 68 chemicals for their abilities to induce Type I spectral changes of P450 2A13 with the Reverse Type I binding spectra observed with P450 1B1: 45 chemicals interacted with both P450s 2A13 and 1B1, indicating that the two enzymes have some similarty of structural features regarding these chemicals. Molecular docking analyses suggest similarities at the active sites of these P450 enzymes. These results indicate that P450 2A13, as well as Family 1 P450 enzymes, is able to catalyze many detoxication and activation reactions with chemicals of environmental interest.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Berne S, Podobnik B, Zupanec N, Novak M, Kraševec N, Turk S, Korošec B, Lah L, Šuligoj E, Stojan J, Gobec S, Komel R. Virtual Screening Yields Inhibitors of Novel Antifungal Drug Target, Benzoate 4-Monooxygenase. J Chem Inf Model 2012; 52:3053-63. [DOI: 10.1021/ci3004418] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sabina Berne
- Faculty of Medicine, Institute
of Biochemistry, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Barbara Podobnik
- Lek Pharmaceuticals d.d., Verovškova 57, SI-1000 Ljubljana, Slovenia
| | - Neja Zupanec
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Metka Novak
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Nada Kraševec
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Samo Turk
- Faculty of Pharmacy, Chair of
Pharmaceutical Chemistry, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Branka Korošec
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Ljerka Lah
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Erika Šuligoj
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Jure Stojan
- Faculty of Medicine, Institute
of Biochemistry, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, Chair of
Pharmaceutical Chemistry, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Radovan Komel
- Faculty of Medicine, Institute
of Biochemistry, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
- Laboratory for Molecular Biology
and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
30
|
Shimada T, Tanaka K, Takenaka S, Murayama N, Martin MV, Foroozesh MK, Yamazaki H, Guengerich FP, Komori M. Structure-function relationships of inhibition of human cytochromes P450 1A1, 1A2, 1B1, 2C9, and 3A4 by 33 flavonoid derivatives. Chem Res Toxicol 2012; 23:1921-35. [PMID: 21053930 DOI: 10.1021/tx100286d] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Structure-function relationships for the inhibition of human cytochrome P450s (P450s) 1A1, 1A2, 1B1, 2C9, and 3A4 by 33 flavonoid derivatives were studied. Thirty-two of the 33 flavonoids tested produced reverse type I binding spectra with P450 1B1, and the potencies of binding were correlated with the abilities to inhibit 7-ethoxyresorufin O-deethylation activity. The presence of a hydroxyl group in flavones, for example, 3-, 5-, and 7-monohydroxy- and 5,7-dihydroxyflavone, decreased the 50% inhibition concentration (IC50) of P450 1B1 from 0.6 μM to 0.09, 0.21, 0.25, and 0.27 μM, respectively, and 3,5,7-trihydroxyflavone (galangin) was the most potent, with an IC50 of 0.003 μM. The introduction of a 4'-methoxy- or 3',4'-dimethoxy group into 5,7-dihydroxyflavone yielded other active inhibitors of P450 1B1 with IC50 values of 0.014 and 0.019 μM, respectively. The above hydroxyl and/or methoxy groups in flavone molecules also increased the inhibition activity with P450 1A1 but not always toward P450 1A2, where 3-, 5-, or 7-hydroxyflavone and 4'-methoxy-5,7-dihydroxyflavone were less inhibitory than flavone itself. P450 2C9 was more inhibited by 7-hydroxy-, 5,7-dihydroxy-, and 3,5,7-trihydroxyflavones than by flavone but was weakly inhibited by 3- and 5-hydroxyflavone. Flavone and several other flavonoids produced type I binding spectra with P450 3A4, but such binding was not always related to the inhibitiory activities toward P450 3A4. These results indicate that there are different mechanisms of inhibition for P450s 1A1, 1A2, 1B1, 2C9, and 3A4 by various flavonoid derivatives and that the number and position of hydroxyl and/or methoxy groups highly influence the inhibitory actions of flavonoids toward these enzymes. Molecular docking studies suggest that there are different mechanisms involved in the interaction of various flavonoids with the active site of P450s, thus causing differences in inhibition of these P450 catalytic activities by flavonoids.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gonzalez J, Marchand-Geneste N, Giraudel JL, Shimada T. Docking and QSAR comparative studies of polycyclic aromatic hydrocarbons and other procarcinogen interactions with cytochromes P450 1A1 and 1B1. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2012; 23:87-109. [PMID: 22150106 DOI: 10.1080/1062936x.2011.636380] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
To obtain chemical clues on the process of bioactivation by cytochromes P450 1A1 and 1B1, some QSAR studies were carried out based on cellular experiments of the metabolic activation of polycyclic aromatic hydrocarbons and heterocyclic aromatic compounds by those enzymes. Firstly, the 3D structures of cytochromes 1A1 and 1B1 were built using homology modelling with a cytochrome 1A2 template. Using these structures, 32 ligands including heterocyclic aromatic compounds, polycyclic aromatic hydrocarbons and corresponding diols, were docked with LigandFit and CDOCKER algorithms. Binding mode analysis highlighted the importance of hydrophobic interactions and the hydrogen bonding network between cytochrome amino acids and docked molecules. Finally, for each enzyme, multilinear regression and artificial neural network QSAR models were developed and compared. These statistical models highlighted the importance of electronic, structural and energetic descriptors in metabolic activation process, and could be used for virtual screening of ligand databases. In the case of P450 1A1, the best model was obtained with artificial neural network analysis and gave an r (2) of 0.66 and an external prediction [Formula: see text] of 0.73. Concerning P450 1B1, artificial neural network analysis gave a much more robust model, associated with an r (2) value of 0.73 and an external prediction [Formula: see text] of 0.59.
Collapse
Affiliation(s)
- J Gonzalez
- Université Bordeaux 1, Talence Cedex, France
| | | | | | | |
Collapse
|
32
|
Sridhar J, Foroozesh M, Stevens CK. QSAR models of cytochrome P450 enzyme 1A2 inhibitors using CoMFA, CoMSIA and HQSAR. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2011; 22:681-697. [PMID: 22004550 PMCID: PMC3371641 DOI: 10.1080/1062936x.2011.623320] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Quantitative structure-activity relationship (QSAR) studies were conducted on an in-house database of cytochrome P450 enzyme 1A2 inhibitors using the comparative molecular field analysis (CoMFA), comparative molecular similarity analysis (CoMSIA) and hologram QSAR (HQSAR) approaches. The database consisted of 36 active molecules featuring varied core structures. The model based on the naphthalene substructure alignment incorporating 19 molecules yielded the best model with a CoMFA cross validation value q(2) of 0.667 and a Pearson correlation coefficient r(2) of 0.976; a CoMSIA q(2) value of 0.616 and r(2) value of 0.985; and a HQSAR q(2) value of 0.652 and r(2) value of 0.917. A second model incorporating 34 molecules aligned using the benzene substructure yielded an acceptable CoMFA model with q(2) value of 0.5 and r(2) value of 0.991. Depending on the core structure of the molecule under consideration, new CYP1A2 inhibitors will be designed based on the results from these models.
Collapse
|
33
|
Shimada T, Murayama N, Tanaka K, Takenaka S, Guengerich FP, Yamazaki H, Komori M. Spectral modification and catalytic inhibition of human cytochromes P450 1A1, 1A2, 1B1, 2A6, and 2A13 by four chemopreventive organoselenium compounds. Chem Res Toxicol 2011; 24:1327-37. [PMID: 21732699 DOI: 10.1021/tx200218u] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Several organoselenium compounds including benzyl selenocyanate (BSC), 1,2-phenylenebis(methylene)selenocyanate (o-XSC), 1,3-phenylenebis(methylene)selenocyanate (m-XSC), and 1,4-phenylenebis(methylene)selenocyanate (p-XSC) have been shown to prevent cancers caused by polycyclic aromatic hydrocarbons (PAHs) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in experimental animals; these chemical carcinogens are activated by human P450 1 and 2A family enzymes, respectively, to carcinogenic metabolites. In this study, we examined whether these selenium compounds interact with and inhibit human P450 1 and 2A enzymes in vitro. Four organoselenium compounds induced reverse Type I binding spectra with P450 1A1, 1A2, and 1B1 and Type I binding spectra with P450 2A6 and 2A13. The spectral dissociation constants (K(s)) for the interaction of P450 1B1 with these chemicals were 3.6-5.7 μM; the values were lower than those with seen with P450 1A1 (19-30 μM) or 1A2 (6.3-13 μM). The K(s) values for Type I binding of P450 2A13 with m-XSC and BSC were both 0.20 μM; the values were very low compared to those for the interaction of P450 2A6 with m-XSC (5.7 μM) and BSC (2.0 μM). Four selenium compounds directly inhibited 7-ethoxyresorufin O-deethylation activities catalyzed by P450 1A1, 1A2, and 1B1 with IC(50) values <1.0 μM, except for the inhibition of P450 1A2 by BSC (1.3 μM). Coumarin 7-hydroxylation activities of P450 2A13 were more inhibited by four selenium compounds than those of P450 2A6, with IC(50) values of 0.22-1.4 μM for P450 2A13 and 2.4-6.2 μM for P450 2A6. Molecular docking studies of the interaction of four organoselenium compounds with human P450 enzymes suggest that these chemicals can be docked into the active sites of these human P450 enzymes and that the sites of the selenocyanate functional groups of these chemicals differ between the P450 1 and 2A family enzymes.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Laboratory of Cellular and Molecular Biology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University , 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Ouellet H, Kells PM, Ortiz de Montellano PR, Podust LM. Reverse type I inhibitor of Mycobacterium tuberculosis CYP125A1. Bioorg Med Chem Lett 2011; 21:332-7. [PMID: 21109436 PMCID: PMC3011832 DOI: 10.1016/j.bmcl.2010.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 11/01/2010] [Indexed: 10/18/2022]
Abstract
Cytochrome P450 CYP125A1 of Mycobacterium tuberculosis, a potential therapeutic target for tuberculosis in humans, initiates degradation of the aliphatic chain of host cholesterol and is essential for establishing M. tuberculosis infection in a mouse model of disease. We explored the interactions of CYP125A1 with a reverse type I inhibitor by X-ray structure analysis and UV-vis spectroscopy. Compound LP10 (α-[(4-methylcyclohexyl)carbonyl amino]-N-4-pyridinyl-1H-indole-3-propanamide), previously identified as a potent type II inhibitor of Trypanosomacruzi CYP51, shifts CYP125A1 to a water-coordinated low-spin state upon binding with low micromolar affinity. When LP10 is present in the active site, the crystal structure and spectral characteristics both demonstrate changes in lipophilic and electronic properties favoring coordination of the iron axial water ligand. These results provide an insight into the structural requirements for developing selective CYP125A1 inhibitors.
Collapse
Affiliation(s)
- Hugues Ouellet
- Department of Pharmaceutical Chemistry and Sandler Center for Drug Discovery, University of California, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
35
|
SINEVA ELENAV, DAVYDOV DMITRIR. Constrained water access to the active site of cytochrome P450 from the piezophilic bacterium Photobacterium profundum. HIGH PRESSURE RESEARCH 2010; 30:466-474. [PMID: 21475616 PMCID: PMC3070315 DOI: 10.1080/08957959.2010.535208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Living species inhabiting ocean deeps must adapt to high hydrostatic pressure. This adaptation, which must enable functioning under conditions of promoted protein hydration, is especially important for proteins such as cytochromes P450 that exhibit functionally important hydration-dehydration dynamics. Here we study the interactions of substrates with cytochrome P450-SS9, a putative fatty acid hydroxylase from the piezophilic bacterium Photobacterium profundum SS9, and characterize the protein's barotropic properties. Comparison of P450-SS9 with cytochrome P450BM-3, a mesophilic fatty acid hydroxylase, suggests that P450-SS9 is characterized by severely confined accessibility and low water occupancy of the active site. This feature may reveal a mechanism of structural adaptation of the piezophilic enzyme. We also demonstrate that saturated and unsaturated fatty acids exert opposite effects on solvent accessibility and hydration of the active site. Modulation of the protein conformation by fatty acids is hypothesized to have an important physiological function in the piezophile.
Collapse
|
36
|
Yasutake Y, Fujii Y, Nishioka T, Cheon WK, Arisawa A, Tamura T. Structural evidence for enhancement of sequential vitamin D3 hydroxylation activities by directed evolution of cytochrome P450 vitamin D3 hydroxylase. J Biol Chem 2010; 285:31193-201. [PMID: 20667833 DOI: 10.1074/jbc.m110.147009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vitamin D(3) hydroxylase (Vdh) isolated from actinomycete Pseudonocardia autotrophica is a cytochrome P450 (CYP) responsible for the biocatalytic conversion of vitamin D(3) (VD(3)) to 1α,25-dihydroxyvitamin D(3) (1α,25(OH)(2)VD(3)) by P. autotrophica. Although its biological function is unclear, Vdh is capable of catalyzing the two-step hydroxylation of VD(3), i.e. the conversion of VD(3) to 25-hydroxyvitamin D(3) (25(OH)VD(3)) and then of 25(OH)VD(3) to 1α,25(OH)(2)VD(3), a hormonal form of VD(3). Here we describe the crystal structures of wild-type Vdh (Vdh-WT) in the substrate-free form and of the highly active quadruple mutant (Vdh-K1) generated by directed evolution in the substrate-free, VD(3)-bound, and 25(OH)VD(3)-bound forms. Vdh-WT exhibits an open conformation with the distal heme pocket exposed to the solvent both in the presence and absence of a substrate, whereas Vdh-K1 exhibits a closed conformation in both the substrate-free and substrate-bound forms. The results suggest that the conformational equilibrium was largely shifted toward the closed conformation by four amino acid substitutions scattered throughout the molecule. The substrate-bound structure of Vdh-K1 accommodates both VD(3) and 25(OH)VD(3) but in an anti-parallel orientation. The occurrence of the two secosteroid binding modes accounts for the regioselective sequential VD(3) hydroxylation activities. Moreover, these structures determined before and after directed evolution, together with biochemical and spectroscopic data, provide insights into how directed evolution has worked for significant enhancement of both the VD(3) 25-hydroxylase and 25(OH)VD(3) 1α-hydroxylase activities.
Collapse
Affiliation(s)
- Yoshiaki Yasutake
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology, Sapporo 062-8517, Japan
| | | | | | | | | | | |
Collapse
|