1
|
Esteves F, Rueff J, Kranendonk M. The Central Role of Cytochrome P450 in Xenobiotic Metabolism-A Brief Review on a Fascinating Enzyme Family. J Xenobiot 2021; 11:94-114. [PMID: 34206277 PMCID: PMC8293344 DOI: 10.3390/jox11030007] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022] Open
Abstract
Human Cytochrome P450 (CYP) enzymes constitute a superfamily of membrane-bound hemoproteins that are responsible for the metabolism of a wide variety of clinically, physiologically, and toxicologically important compounds. These heme-thiolate monooxygenases play a pivotal role in the detoxification of xenobiotics, participating in the metabolism of many structurally diverge compounds. This short-review is intended to provide a summary on the major roles of CYPs in Phase I xenobiotic metabolism. The manuscript is focused on eight main topics that include the most relevant aspects of past and current CYP research. Initially, (I) a general overview of the main aspects of absorption, distribution, metabolism, and excretion (ADME) of xenobiotics are presented. This is followed by (II) a background overview on major achievements in the past of the CYP research field. (III) Classification and nomenclature of CYPs is briefly reviewed, followed by (IV) a summary description on CYP's location and function in mammals. Subsequently, (V) the physiological relevance of CYP as the cornerstone of Phase I xenobiotic metabolism is highlighted, followed by (VI) reviewing both genetic determinants and (VI) nongenetic factors in CYP function and activity. The last topic of the review (VIII) is focused on the current challenges of the CYP research field.
Collapse
Affiliation(s)
- Francisco Esteves
- Center for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Huma Toxicology, NOVA Medical School/Faculty of Medical Sciences, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.R.); (M.K.)
| | | | | |
Collapse
|
2
|
Ming X, Michaelson-Richie ED, Groehler AS, Villalta PW, Campbell C, Tretyakova NY. Cross-linking of the DNA repair protein O 6-alkylguanine DNA alkyltransferase to DNA in the presence of cisplatin. DNA Repair (Amst) 2020; 89:102840. [PMID: 32283495 DOI: 10.1016/j.dnarep.2020.102840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/23/2022]
Abstract
1,1,2,2-cis-diamminedichloroplatinum (II) (cisplatin) is a chemotherapeutic agent widely used in the clinic to treat various cancers. The antitumor activity of cisplatin is generally attributed to its ability to form intrastrand and interstrand DNA-DNA cross-links via sequential platination of two nucleophilic sites within the DNA duplex. However, cisplatin also induces DNA- protein lesions (DPCs) that may contribute to its biological effects due to their ability to block DNA replication and transcription. We previously reported that over 250 nuclear proteins including high mobility group proteins, histone proteins, and elongation factors formed DPCs in human HT1080 cells treated with cisplatin (Ming et al. Chem. Res. Toxicol. 2017, 30, 980-995). Interestingly, cisplatin-induced DNA-protein conjugates were reversed upon heating, by an unknown mechanism. In the present work, DNA repair protein O6-alkylguanine DNA alkyltransferase (AGT) was used as a model to investigate the molecular details of cisplatin-mediated DNA-protein cross-linking and to establish the mechanism of their reversal. We found that AGT is readily cross-linked to DNA in the presence of cisplatin. HPLC-ESI+-MS/MS sequencing of tryptic peptides originating from dG-Pt-AGT complexes revealed that the cross-linking occurred at six sites within this protein including Glu110, Lys125, Cys145, His146, Arg147, and Cys150. Cisplatin-induced Lys-Gua cross-links (1,1-cis-diammine-2-(5-amino-5-carboxypentyl)amino-2-(2'-deoxyguanosine-7-yl)-platinum(II) (dG-Pt-Lys) were detected by HPLC-ESI+-MS/MS of total digests of modified protein in comparison with the corresponding authentic standard. Upon heating, dG-Pt-AGT complexes were subject to platination migration from protein to DNA, forming cis-[Pt(NH3)2{d(GpG)}] cross-links which were detected by HPLC-ESI+-MS/MS. Our results provide a new insight into the mechanism of cisplatin-mediated DNA-protein cross-linking and their dynamic equilibrium with the corresponding DNA-DNA lesions.
Collapse
Affiliation(s)
- Xun Ming
- Department of Medicinal Chemistry and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Erin D Michaelson-Richie
- Department of Medicinal Chemistry and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Arnold S Groehler
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Peter W Villalta
- Mass Spectrometry Core at the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Colin Campbell
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Natalia Y Tretyakova
- Department of Medicinal Chemistry and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Lassila T, Rousu T, Mattila S, Chesné C, Pelkonen O, Turpeinen M, Tolonen A. Formation of GSH-trapped reactive metabolites in human liver microsomes, S9 fraction, HepaRG-cells, and human hepatocytes. J Pharm Biomed Anal 2015; 115:345-51. [PMID: 26263063 DOI: 10.1016/j.jpba.2015.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/19/2015] [Accepted: 07/21/2015] [Indexed: 12/22/2022]
|
4
|
Sinha S, Ahire D, Wagh S, Mullick D, Sistla R, Selvakumar K, Cortes JC, Putlur SP, Mandlekar S, Johnson BM. Electrophilicity of pyridazine-3-carbonitrile, pyrimidine-2-carbonitrile, and pyridine-carbonitrile derivatives: a chemical model to describe the formation of thiazoline derivatives in human liver microsomes. Chem Res Toxicol 2014; 27:2052-61. [PMID: 25372409 DOI: 10.1021/tx500256j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Certain aromatic nitriles are well-known inhibitors of cysteine proteases. The mode of action of these compounds involves the formation of a reversible or irreversible covalent bond between the nitrile and a thiol group in the active site of the enzyme. However, the reactivity of these aromatic nitrile-substituted heterocycles may lead inadvertently to nonspecific interactions with DNA, protein, glutathione, and other endogenous components, resulting in toxicity and complicating the use of these compounds as therapeutic agents. In the present study, the intrinsic reactivity and associated structure-property relationships of cathepsin K inhibitors featuring substituted pyridazines [6-phenylpyridazine-3-carbonitrile, 6-(4-fluorophenyl)pyridazine-3-carbonitrile, 6-(4-methoxyphenyl)pyridazine-3-carbonitrile, 6-p-tolylpyridazine-3-carbonitrile], pyrimidines [5-p-tolylpyrimidine-2-carbonitrile, 5-(4-fluorophenyl)pyrimidine-2-carbonitrile], and pyridines [5-p-tolylpicolinonitrile and 5-(4-fluorophenyl)picolinonitrile] were evaluated using a combination of computational and analytical approaches to establish correlations between electrophilicity and levels of metabolites that were formed in glutathione- and N-acetylcysteine-supplemented human liver microsomes. Metabolites that were characterized in this study featured substituted thiazolines that were formed following rearrangements of transient glutathione and N-acetylcysteine conjugates. Peptidases including γ-glutamyltranspeptidase were shown to catalyze the formation of these products, which were formed to lesser extents in the presence of the selective γ-glutamyltranspeptidase inhibitor acivicin and the nonspecific peptidase inhibitors phenylmethylsulfonyl fluoride and aprotinin. Of the chemical series mentioned above, the pyrimidine series was the most susceptible to metabolism to thiazoline-containing products, followed, in order, by the pyridazine and pyridine series. This trend was in keeping with the diminishing electrophilicity across these series, as demonstrated by in silico modeling. Hence, mechanistic insights gained from this study could be used to assist a medicinal chemistry campaign to design cysteine protease inhibitors that were less prone to the formation of covalent adducts.
Collapse
Affiliation(s)
- Sarmistha Sinha
- Pharmaceutical Candidate Optimization, ‡Medicinal Chemistry Department, and §Advanced Biotechnology Department, Biocon Bristol-Myers Squibb R&D Center (BBRC), Syngene International Ltd , Plot No. 2 & 3, Bommasandra IV Phase, Jigani Link Road, Bangalore 560100, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Diclofenac toxicity in human intestine ex vivo is not related to the formation of intestinal metabolites. Arch Toxicol 2014; 89:107-19. [PMID: 24770551 DOI: 10.1007/s00204-014-1242-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 04/10/2014] [Indexed: 12/22/2022]
Abstract
The use of diclofenac (DCF), a nonsteroidal anti-inflammatory drug, is associated with a high prevalence of gastrointestinal side effects. In vivo studies in rodents suggested that reactive metabolites of DCF produced by the liver or the intestine might be responsible for this toxicity. In the present study, precision-cut intestinal slices (PCIS) prepared from the jejunum of 18 human donors were used as an ex vivo model to investigate whether DCF intestinal metabolites are responsible for its intestinal toxicity in man. PCIS were incubated with a concentration range of DCF (0-600 µM) up to 24 h. DCF (≥400 µM) caused direct toxicity to the intestine as demonstrated by ATP depletion, morphological damage, caspase 3 activation, and lactate dehydrogenase leakage. Three main metabolites produced by PCIS (4'-hydroxy DCF, 5-hydroxy DCF, and DCF acyl glucuronide) were detected by HPLC. Protein adducts were detected by immunohistochemical staining and showed correlation with the intestinal metabolites. DCF induced similar toxicity to each of the samples regardless of the variation in metabolism among them. Less metabolites were produced by slices incubated with 400 µM DCF than with 100 µM DCF. The addition of the metabolic inhibitors such as ketoconazole, cimetidine, or borneol decreased the metabolite formation but increased the toxicity. The results suggest that DCF can induce intestinal toxicity in human PCIS directly at therapeutically relevant concentrations, independent of the reactive metabolites 4'-OH DCF, 5-OH DCF, or diclofenac acylglucuronide produced by the liver or formed in the intestine.
Collapse
|
6
|
Wickramaratne S, Tretyakova NY. Structure elucidation of DNA-protein crosslinks by using reductive desulfurization and liquid chromatography-tandem mass spectrometry. Chembiochem 2014; 15:353-5. [PMID: 24436288 DOI: 10.1002/cbic.201300757] [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: 12/02/2013] [Indexed: 11/12/2022]
Abstract
Easier with ethyl: Guengerich and co-workers have developed a powerful new approach to the structure elucidation of hydrolytically stable AGT-DNA crosslinks by reductive desulfurization of the thioether linkage between AGT and DNA to convert cysteine DPCs to the corresponding ethyl-DNA adducts, which can be readily characterized by LC-MSn.
Collapse
Affiliation(s)
- Susith Wickramaratne
- University of Minnesota Masonic Cancer Center and the Department of Chemistry, 2231 6th Street SE, Room 2-220 CCRB, Minneapolis, MN 55455 (USA)
| | | |
Collapse
|
7
|
Zhao L, Balbo S, Wang M, Upadhyaya P, Khariwala SS, Villalta PW, Hecht SS. Quantitation of pyridyloxobutyl-DNA adducts in tissues of rats treated chronically with (R)- or (S)-N'-nitrosonornicotine (NNN) in a carcinogenicity study. Chem Res Toxicol 2013; 26:1526-35. [PMID: 24001146 PMCID: PMC3848204 DOI: 10.1021/tx400235x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We quantified DNA adducts resulting from 2'-hydroxylation of enantiomers of the tobacco-specific nitrosamine N'-nitrosonornicotine (NNN) in tissues of male F-344 rats after 10, 30, 50, and 70 weeks of treatment with 14 ppm in the drinking water. These rats were in subgroups of a carcinogenicity study in which (S)-NNN was highly tumorigenic in the oral cavity and esophagus, while (R)-NNN was relatively weakly active. DNA adducts were quantified by liquid chromatography-electrospray ionization-tandem mass spectrometry in six tissues: oral mucosa, esophageal mucosa, nasal respiratory mucosa, nasal olfactory mucosa, liver, and lung. O²-[4-(3-Pyridyl)-4-oxobut-1-yl]thymidine (O²-POB-dThd, 7) and 7-[4-(3-pyridyl)-4-oxobut-1-yl]-2'-deoxyguanosine (7-POB-dGuo, 8), the latter as 7-[4-(3-pyridyl)-4-oxobut-1-yl]guanine (7-POB-Gua, 11), were detected at each time point in each tissue. In the target tissues for carcinogenicity, oral mucosa and esophageal mucosa, levels of 7-POB-Gua (11) and O²-POB-dThd (7) were similar, or 11 predominated, while in all other tissues at all time points for both enantiomers, 7 was clearly present in greater amounts than 11. Total measured DNA adduct levels in esophageal mucosa and oral mucosa were higher in rats treated with (S)-NNN than (R)-NNN. The highest adduct levels were found in the nasal respiratory mucosa. DNA adducts generally persisted in all tissues without any sign of substantial decreases throughout the 70 week time course. The results of this study suggest that inefficient repair of 7-POB-dGuo (8) in the rat oral cavity and esophagus may be important in carcinogenesis by NNN and support the development of these DNA adducts as potential biomarkers of NNN metabolic activation in people who use tobacco products.
Collapse
Affiliation(s)
- Lijiao Zhao
- College of Life Science and Bioengineering, Beijing University of Technology , Beijing 100124, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Tretyakova NY, Michaelson-Richie ED, Gherezghiher TB, Kurtz J, Ming X, Wickramaratne S, Campion M, Kanugula S, Pegg AE, Campbell C. DNA-reactive protein monoepoxides induce cell death and mutagenesis in mammalian cells. Biochemistry 2013; 52:3171-81. [PMID: 23566219 DOI: 10.1021/bi400273m] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although cytotoxic alkylating agents possessing two electrophilic reactive groups are thought to act by cross-linking cellular biomolecules, their exact mechanisms of action have not been established. In cells, these compounds form a mixture of DNA lesions, including nucleobase monoadducts, interstrand and intrastrand cross-links, and DNA-protein cross-links (DPCs). Interstrand DNA-DNA cross-links block replication and transcription by preventing DNA strand separation, contributing to toxicity and mutagenesis. In contrast, potential contributions of drug-induced DPCs are poorly understood. To gain insight into the biological consequences of DPC formation, we generated DNA-reactive protein reagents and examined their toxicity and mutagenesis in mammalian cells. Recombinant human O(6)-alkylguanine DNA alkyltransferase (AGT) protein or its variants (C145A and K125L) were treated with 1,2,3,4-diepoxybutane to yield proteins containing 2-hydroxy-3,4-epoxybutyl groups on cysteine residues. Gel shift and mass spectrometry experiments confirmed that epoxide-functionalized AGT proteins formed covalent DPC but no other types of nucleobase damage when incubated with duplex DNA. Introduction of purified AGT monoepoxides into mammalian cells via electroporation generated AGT-DNA cross-links and induced cell death and mutations at the hypoxanthine-guanine phosphoribosyltransferase gene. Smaller numbers of DPC lesions and reduced levels of cell death were observed when using protein monoepoxides generated from an AGT variant that fails to accumulate in the cell nucleus (K125L), suggesting that nuclear DNA damage is required for toxicity. Taken together, these results indicate that AGT protein monoepoxides produce cytotoxic and mutagenic DPC lesions within chromosomal DNA. More generally, these data suggest that covalent DPC lesions contribute to the cytotoxic and mutagenic effects of bis-electrophiles.
Collapse
Affiliation(s)
- Natalia Y Tretyakova
- Department of Medicinal Chemistry and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Naticchia MR, Brown HA, Garcia FJ, Lamade AM, Justice SL, Herrin RP, Morano KA, West JD. Bifunctional electrophiles cross-link thioredoxins with redox relay partners in cells. Chem Res Toxicol 2013; 26:490-7. [PMID: 23414292 DOI: 10.1021/tx4000123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thioredoxin protects cells against oxidative damage by reducing disulfide bonds in improperly oxidized proteins. Previously, we found that the baker's yeast cytosolic thioredoxin Trx2 undergoes cross-linking to form several protein-protein complexes in cells treated with the bifunctional electrophile divinyl sulfone (DVSF). Here, we report that the peroxiredoxin Tsa1 and the thioredoxin reductase Trr1, both of which function in a redox relay network with thioredoxin, become cross-linked in complexes with Trx2 upon DVSF treatment. Treatment of yeast with other bifunctional electrophiles, including diethyl acetylenedicarboxylate (DAD), mechlorethamine (HN2), and 1,2,3,4-diepoxybutane (DEB), resulted in the formation of similar cross-linked complexes. Cross-linking of Trx2 and Tsa1 to other proteins by DVSF and DAD is dependent on modification of the active site Cys residues within these proteins. In addition, the human cytosolic thioredoxin, cytosolic thioredoxin reductase, and peroxiredoxin 2 form cross-linked complexes to other proteins in the presence of DVSF, although each protein shows different susceptibilities to modification by DAD, HN2, and DEB. Taken together, our results indicate that bifunctional electrophiles potentially disrupt redox homeostasis in yeast and human cells by forming cross-linked complexes between thioredoxins and their redox partners.
Collapse
Affiliation(s)
- Matthew R Naticchia
- Biochemistry and Molecular Biology Program, Departments of Biology and Chemistry, The College of Wooster, Wooster, Ohio 44691, United States
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Ballard P, Brassil P, Bui KH, Dolgos H, Petersson C, Tunek A, Webborn PJH. The right compound in the right assay at the right time: an integrated discovery DMPK strategy. Drug Metab Rev 2012; 44:224-52. [DOI: 10.3109/03602532.2012.691099] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
11
|
Njuguna NM, Masimirembwa C, Chibale K. Identification and characterization of reactive metabolites in natural products-driven drug discovery. JOURNAL OF NATURAL PRODUCTS 2012; 75:507-513. [PMID: 22296642 DOI: 10.1021/np200786j] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Toxicity of natural products arising from their metabolic biotransformation into reactive chemical intermediates is an important reason for high attrition rates in early drug discovery efforts. Screening promising natural products for their likelihood to form such metabolites is therefore an important step in identifying potential liabilities in the drug development process. However, such screening is complicated by the need to have test methods that are sensitive, reliable, accurate, efficient, and cost-effective enough to allow for routine identification and characterization of the reactive metabolites. These metabolites are typically formed in minute quantities, usually through minor metabolic pathways, and, due to their highly reactive and therefore transient chemical nature, pose considerable analytical challenges in attempts to determine their properties. Understanding the formation of reactive metabolites may be used as the basis for synthetic chemical modification of parent natural products aimed at bypassing such harmful bioactivation. This paper highlights the general principles and protocols commonly used to predict and study the formation of reactive metabolites in vitro and how the data obtained from such studies can be used in the development of safer drugs from natural products.
Collapse
Affiliation(s)
- Nicholas M Njuguna
- Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | | | | |
Collapse
|
12
|
Abstract
'It is better to be useful than perfect'. This review attempts to critically cover and assess the currently available approaches and tools to answer the crucial question: Is it possible (and if it is, to what extent is it possible) to predict in vivo metabolites and their abundances on the basis of in vitro and preclinical animal studies? In preclinical drug development, it is possible to produce metabolite patterns from a candidate drug by virtual means (i.e., in silico models), but these are not yet validated. However, they may be useful to cover the potential range of metabolites. In vitro metabolite patterns and apparent relative abundances are produced by various in vitro systems employing tissue preparations (mainly liver) and in most cases using liquid chromatography-mass spectrometry analytical techniques for tentative identification. The pattern of the metabolites produced depends on the enzyme source; the most comprehensive source of drug-metabolizing enzymes is cultured human hepatocytes, followed by liver homogenate fortified with appropriate cofactors. For specific purposes, such as the identification of metabolizing enzyme(s), recombinant enzymes can be used. Metabolite data from animal in vitro and in vivo experiments, despite known species differences, may help pinpoint metabolites that are not apparently produced in in vitro human systems, or suggest alternative experimental approaches. The range of metabolites detected provides clues regarding the enzymes attacking the molecule under study. We also discuss established approaches to identify the major enzymes. The last question, regarding reliability and robustness of metabolite extrapolations from in vitro to in vivo, both qualitatively and quantitatively, cannot be easily answered. There are a number of examples in the literature suggesting that extrapolations are generally useful, but there are only a few systematic and comprehensive studies to validate in vitro-in vivo extrapolations. In conclusion, extrapolation from preclinical metabolite data to the in vivo situation is certainly useful, but it is not known to what extent.
Collapse
|
13
|
Vijayalakshmi KP, Mohan N, Ajitha MJ, Suresh CH. Mechanism of epoxide hydrolysis in microsolvated nucleotide bases adenine, guanine and cytosine: a DFT study. Org Biomol Chem 2011; 9:5115-22. [PMID: 21629892 DOI: 10.1039/c1ob05093a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Six water molecules have been used for microsolvation to outline a hydrogen bonded network around complexes of ethylene epoxide with nucleotide bases adenine (EAw), guanine (EGw) and cytosine (ECw). These models have been developed with the MPWB1K-PCM/6-311++G(3df,2p)//MPWB1K/6-31+G(d,p) level of DFT method and calculated S(N)2 type ring opening of the epoxide due to amino group of the nucleotide bases, viz. the N6 position of adenine, N2 position of guanine and N4 position of cytosine. Activation energy (E(act)) for the ring opening was found to be 28.06, 28.64, and 28.37 kcal mol(-1) respectively for EAw, EGw and ECw. If water molecules were not used, the reactions occurred at considerably high value of E(act), viz. 53.51 kcal mol(-1) for EA, 55.76 kcal mol(-1) for EG and 56.93 kcal mol(-1) for EC. The ring opening led to accumulation of negative charge on the developing alkoxide moiety and the water molecules around the charge localized regions showed strong hydrogen bond interactions to provide stability to the intermediate systems EAw-1, EGw-1 and ECw-1. This led to an easy migration of a proton from an activated water molecule to the alkoxide moiety to generate a hydroxide. Almost simultaneously, a proton transfer chain reaction occurred through the hydrogen bonded network of water molecules and resulted in the rupture of one of the N-H bonds of the quaternized amino group. The highest value of E(act) for the proton transfer step of the reaction was 2.17 kcal mol(-1) for EAw, 2.93 kcal mol(-1) for EGw and 0.02 kcal mol(-1) for ECw. Further, the overall reaction was exothermic by 17.99, 22.49 and 13.18 kcal mol(-1) for EAw, EGw and ECw, respectively, suggesting that the reaction is irreversible. Based on geometric features of the epoxide-nucleotide base complexes and the energetics, the highest reactivity is assigned for adenine followed by cytosine and guanine. Epoxide-mediated damage of DNA is reported in the literature and the present results suggest that hydrated DNA bases become highly S(N)2 active on epoxide systems and the occurrence of such reactions can inflict permanent damage to the DNA.
Collapse
Affiliation(s)
- Kunduchi P Vijayalakshmi
- Computational Modeling and Simulation Section, National Institute for Interdisciplinary Science and Technology (CSIR), Trivandrum, 695 019, India
| | | | | | | |
Collapse
|
14
|
Rousu T, Tolonen A. Characterization of cyanide-trapped methylated metabonates formed during reactive drug metabolite screening in vitro. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2011; 25:1382-1390. [PMID: 21504003 DOI: 10.1002/rcm.5005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Reactive metabolites are estimated to be one of the main reasons behind unexpected drug-induced toxicity, by binding covalently to cell proteins or DNA. Due to their high reactivity and short lifespan, reactive metabolites are analyzed after chemical trapping with nucleophilic agents such as glutathione or cyanide. Recently, unexplained and uncharacterized methylated reaction products were reported in a human liver microsome based reactive metabolite trapping assay utilizing potassium cyanide as a trapping agent. Here, a similar assay was utilized to produce mono- or dimethylated and further cyanide-trapped reaction products from propranolol, amlodipine and ciprofloxacin, followed by ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC/TOF-MS) and ultra-performance liquid chromatography/tandem mass spectrometry (UPLC/MS/MS) experiments for their more detailed structural elucidation. Formation of all observed cyanide-trapped products was clearly NADPH-dependent and thus metabolism-mediated. The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. As the methylation reaction was suggested to be involved in formation of the actual reactive iminium ion, the observed cyanide-trapped products were experimental artifacts rather than trapped reactive metabolites. The results stress that to avoid overestimating the formation of reactive metabolites in vitro, this methylation phenomenon should be taken into account when interpreting the results of cyanide-utilizing reactive metabolite trapping assays. This in turn emphasizes the importance of identification of the observed cyano conjugates during such studies. Yet, metabolite identification has a high importance to avoid overestimation of in vitro metabolic clearance in the cases where this kind of metabonate formation has a high impact in the disappearance rate of the compound.
Collapse
Affiliation(s)
- Timo Rousu
- Department of Chemistry, University of Oulu, P.O. Box 3000, 90014 Oulu, Finland
| | | |
Collapse
|
15
|
Pegg AE. Multifaceted roles of alkyltransferase and related proteins in DNA repair, DNA damage, resistance to chemotherapy, and research tools. Chem Res Toxicol 2011; 24:618-39. [PMID: 21466232 DOI: 10.1021/tx200031q] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) is a widely distributed, unique DNA repair protein that acts as a single agent to directly remove alkyl groups located on the O(6)-position of guanine from DNA restoring the DNA in one step. The protein acts only once, and its alkylated form is degraded rapidly. It is a major factor in counteracting the mutagenic, carcinogenic, and cytotoxic effects of agents that form such adducts including N-nitroso-compounds and a number of cancer chemotherapeutics. This review describes the structure, function, and mechanism of action of AGTs and of a family of related alkyltransferase-like proteins, which do not act alone to repair O(6)-alkylguanines in DNA but link repair to other pathways. The paradoxical ability of AGTs to stimulate the DNA-damaging ability of dihaloalkanes and other bis-electrophiles via the formation of AGT-DNA cross-links is also described. Other important properties of AGTs include the ability to provide resistance to cancer therapeutic alkylating agents, and the availability of AGT inhibitors such as O(6)-benzylguanine that might overcome this resistance is discussed. Finally, the properties of fusion proteins in which AGT sequences are linked to other proteins are outlined. Such proteins occur naturally, and synthetic variants engineered to react specifically with derivatives of O(6)-benzylguanine are the basis of a valuable research technique for tagging proteins with specific reagents.
Collapse
Affiliation(s)
- Anthony E Pegg
- Department of Cellular and Molecular Physiology, Milton S. Hershey Medical Center, Pennsylvania State University College of Medicine , Pennsylvania 17033, United States.
| |
Collapse
|
16
|
Schwöbel JAH, Koleva YK, Enoch SJ, Bajot F, Hewitt M, Madden JC, Roberts DW, Schultz TW, Cronin MTD. Measurement and Estimation of Electrophilic Reactivity for Predictive Toxicology. Chem Rev 2011; 111:2562-96. [DOI: 10.1021/cr100098n] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Johannes A. H. Schwöbel
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Yana K. Koleva
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Steven J. Enoch
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Fania Bajot
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Mark Hewitt
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Judith C. Madden
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - David W. Roberts
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| | - Terry W. Schultz
- College of Veterinary Medicine, Department of Comparative Medicine, The University of Tennessee, 2407 River Drive, Knoxville, Tennessee 37996-4543, United States
| | - Mark T. D. Cronin
- School of Pharmacy and Chemistry, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England
| |
Collapse
|
17
|
Kalgutkar AS, Mascitti V, Sharma R, Walker GW, Ryder T, McDonald TS, Chen Y, Preville C, Basak A, McClure KF, Kohrt JT, Robinson RP, Munchhof MJ, Cornelius P. Intrinsic Electrophilicity of a 4-Substituted-5-cyano-6-(2-methylpyridin-3-yloxy)pyrimidine Derivative: Structural Characterization of Glutathione Conjugates in Vitro. Chem Res Toxicol 2011; 24:269-78. [DOI: 10.1021/tx100429x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Amit S. Kalgutkar
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Vincent Mascitti
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Raman Sharma
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory W. Walker
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Tim Ryder
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Thomas S. McDonald
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yue Chen
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Cathy Preville
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Arindrajit Basak
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kim F. McClure
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jeffrey T. Kohrt
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ralph P. Robinson
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael J. Munchhof
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Peter Cornelius
- Pharmacokinetics, Dynamics, and Metabolism Department, ‡Medicinal Chemistry Department and §Cardiovascular and Metabolic Diseases Department, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
18
|
Michaelson-Richie ED, Loeber RL, Codreanu SG, Ming X, Liebler DC, Campbell C, Tretyakova NY. DNA-protein cross-linking by 1,2,3,4-diepoxybutane. J Proteome Res 2011; 9:4356-67. [PMID: 20666492 DOI: 10.1021/pr1000835] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
1,2,3,4-diepoxybutane (DEB) is a strongly genotoxic diepoxide hypothesized to be the ultimate carcinogenic metabolite of the common industrial chemical and environmental carcinogen 1,3-butadiene. DEB is a bis-electrophile capable of cross-linking cellular biomolecules to form DNA-DNA and DNA-protein cross-links (DPCs), which are thought to play a central role in its biological activity. Previous studies with recombinant proteins have shown that the biological outcomes of DEB-induced DPCs are strongly influenced by protein identities. The present work combines affinity capture methodology with mass spectrometry-based proteomics and immunological detection to identify the proteins that form DPCs in nuclear extracts from human cervical carcinoma (HeLa) cells. We identified 39 human proteins that form covalent DPCs in the presence of DEB. DNA-protein cross-linking efficiency following treatment with 25 mM DEB was 2-12%, depending on protein identity. High-performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI+-MS/MS) analysis of the total proteolytic digests of cross-linked proteins revealed the presence of 1-(S-cysteinyl)-4-(guan-7-yl)-2,3-butanediol conjugates, suggesting that DEB forms DPCs between cysteine thiols within proteins and the N-7 guanine positions within DNA.
Collapse
Affiliation(s)
- Erin D Michaelson-Richie
- Department of Medicinal Chemistry and the Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
van der Kolk JH, Wijnberg ID, Westermann CM, Dorland L, de Sain-van der Velden MGM, Kranenburg LC, Duran M, Dijkstra JA, van der Lugt JJ, Wanders RJA, Gruys E. Equine acquired multiple acyl-CoA dehydrogenase deficiency (MADD) in 14 horses associated with ingestion of Maple leaves (Acer pseudoplatanus) covered with European tar spot (Rhytisma acerinum). Mol Genet Metab 2010; 101:289-91. [PMID: 20655779 DOI: 10.1016/j.ymgme.2010.06.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 06/27/2010] [Accepted: 06/27/2010] [Indexed: 11/30/2022]
Abstract
This case-series describes fourteen horses suspected of equine acquired multiple acyl-CoA dehydrogenase deficiency (MADD) also known as atypical myopathy of which seven cases were confirmed biochemically with all horses having had access to leaves of the Maple tree (Acer pseudoplatanus) covered with European tar spot (Rhytisma acerinum). Assessment of organic acids, glycine conjugates, and acylcarnitines in urine was regarded as gold standard in the biochemical diagnosis of equine acquired multiple acyl-CoA dehydrogenase deficiency.
Collapse
Affiliation(s)
- J H van der Kolk
- Department of Equine Sciences, Medicine Section, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Huang R, Southall N, Xia M, Cho MH, Jadhav A, Nguyen DT, Inglese J, Tice RR, Austin CP. Weighted feature significance: a simple, interpretable model of compound toxicity based on the statistical enrichment of structural features. Toxicol Sci 2009; 112:385-93. [PMID: 19805409 PMCID: PMC2777082 DOI: 10.1093/toxsci/kfp231] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 09/15/2009] [Indexed: 11/14/2022] Open
Abstract
In support of the U.S. Tox21 program, we have developed a simple and chemically intuitive model we call weighted feature significance (WFS) to predict the toxicological activity of compounds, based on the statistical enrichment of structural features in toxic compounds. We trained and tested the model on the following: (1) data from quantitative high-throughput screening cytotoxicity and caspase activation assays conducted at the National Institutes of Health Chemical Genomics Center, (2) data from Salmonella typhimurium reverse mutagenicity assays conducted by the U.S. National Toxicology Program, and (3) hepatotoxicity data published in the Registry of Toxic Effects of Chemical Substances. Enrichments of structural features in toxic compounds are evaluated for their statistical significance and compiled into a simple additive model of toxicity and then used to score new compounds for potential toxicity. The predictive power of the model for cytotoxicity was validated using an independent set of compounds from the U.S. Environmental Protection Agency tested also at the National Institutes of Health Chemical Genomics Center. We compared the performance of our WFS approach with classical classification methods such as Naive Bayesian clustering and support vector machines. In most test cases, WFS showed similar or slightly better predictive power, especially in the prediction of hepatotoxic compounds, where WFS appeared to have the best performance among the three methods. The new algorithm has the important advantages of simplicity, power, interpretability, and ease of implementation.
Collapse
Affiliation(s)
- Ruili Huang
- Department of Health and Human Services, NIH Chemical Genomics Center, National Institutes of Health, Bethesda, Maryland 20892-3370, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Josephy PD, Kent M, Mannervik B. Single-nucleotide polymorphic variants of human glutathione transferase T1-1 differ in stability and functional properties. Arch Biochem Biophys 2009; 490:24-9. [DOI: 10.1016/j.abb.2009.07.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 07/30/2009] [Accepted: 07/31/2009] [Indexed: 02/07/2023]
|
22
|
Kalapila AG, Loktionova NA, Pegg AE. Effect of O6-alkylguanine-DNA alkyltransferase on genotoxicity of epihalohydrins. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2009; 50:502-514. [PMID: 19472322 PMCID: PMC2855547 DOI: 10.1002/em.20491] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The effect of O(6)-alkylguanine-DNA alkyltransferase (AGT) on the toxicity and mutagenicity of epihalohydrins was studied. AGT is a DNA repair protein that protects cells from agents that produce genotoxic O(6)-alkylguanine lesions by transferring the alkyl group to an internal cysteine residue (Cys(145) in human AGT) in a single-step. This cysteine acceptor site is highly reactive and epihalohydrins reacted readily with AGT at this site with a halide order of reactivity of Br > Cl > F. AGT expression in bacterial cells caused a very large increase in the mutagenicity and cytotoxicity of epibromohydrin. The mutations were almost all G:C to A:T transitions. Epichlorohydrin also augmented AGT-mediated mutagenesis but to a lesser extent than epibromohydrin. In vitro experiments showed that AGT was covalently cross-linked to DNA in the presence of epibromohydrin and that this conjugation occurred predominantly at Cys(145), and to a smaller extent at Cys(150), a less reactive residue also located within the active site pocket. Two pathways yielding the AGT-DNA adduct were found to occur. The predominant mechanism results in an AGT-epihalohydrin intermediate, which, facilitated by the DNA binding properties of AGT, then reacts covalently with DNA. The second pathway involves an initial reactive DNA-epihalohydrin intermediate that subsequently reacts with AGT. Our results show that the paradoxical AGT-mediated increase in genotoxicity which has previously been shown to occur with dihaloalkanes, butadiene diepoxide and nitrogen mustards, also occurs with epihalohydrins and is likely to contribute to their toxicity and mutagenicity.
Collapse
Affiliation(s)
| | | | - Anthony E. Pegg
- Correspondence to: Anthony E. Pegg, Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033.
| |
Collapse
|
23
|
Loecken EM, Dasari S, Hill S, Tabb DL, Guengerich FP. The bis-electrophile diepoxybutane cross-links DNA to human histones but does not result in enhanced mutagenesis in recombinant systems. Chem Res Toxicol 2009; 22:1069-76. [PMID: 19364102 PMCID: PMC2696559 DOI: 10.1021/tx900037u] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1,2-Dibromoethane and 1,3-butadiene are cancer suspects present in the environment and have been used widely in industry. The mutagenic properties of 1,2-dibromoethane and the 1,3-butadiene oxidation product diepoxybutane are thought to be related to the bis-electrophilic character of these chemicals. The discovery that overexpression of O(6)-alkylguanine alkyltransferase (AGT) enhances bis-electrophile-induced mutagenesis prompted a search for other proteins that may act by a similar mechanism. A human liver screen for nuclear proteins that cross-link with DNA in the presence of 1,2-dibromoethane identified histones H2b and H3 as candidate proteins. Treatment of isolated histones H2b and H3 with diepoxybutane resulted in DNA-protein cross-links and produced protein adducts, and DNA-histone H2b cross-links were identified (immunochemically) in Escherichia coli cells expressing histone H2b. However, heterologous expression of histone H2b in E. coli failed to enhance bis-electrophile-induced mutagenesis. These results are similar to those found with the cross-link candidate glyceraldehyde 3-phosphate dehydrogenase (GAPDH) [ Loecken , E. M. and Guengerich , F. P. ( 2008 ) Chem. Res. Toxicol. 21 , 453 - 458 ], but in contrast to GAPDH, histone H2b bound DNA with even higher affinity than AGT. The extent of DNA cross-linking of isolated histone H2b was similar to that of AGT, suggesting that differences in postcross-linking events explain the difference in mutagenesis.
Collapse
Affiliation(s)
| | | | - Salisha Hill
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232−0146 and Department of Biomedical Informatics, and the Proteomics Laboratory of the Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | | | - F. Peter Guengerich
- Address correspondence to: Prof. F. Peter Guengerich Department of Biochemistry and Center in Molecular Toxicology Vanderbilt University School of Medicine 638 Robinson Research Building 2200 Pierce Avenue Nashville, Tennessee 37232−0146 Telephone: (615) 322−2261 FAX: (615) 322−3141 E-mail:
| |
Collapse
|
24
|
Rousu T, Pelkonen O, Tolonen A. Rapid detection and characterization of reactive drug metabolites in vitro using several isotope-labeled trapping agents and ultra-performance liquid chromatography/time-of-flight mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2009; 23:843-855. [PMID: 19224530 DOI: 10.1002/rcm.3953] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Reactive metabolites are believed to be one of the main reasons for unexpected drug-induced toxicity issues, by forming covalent adducts with cell proteins or DNA. Due to their high reactivity and short lifespan they are not directly detected by traditional analytical methods, but are most traditionally analyzed by liquid chromatography/tandem mass spectrometry (LC/MS/MS) after chemical trapping with nucleophilic agents such as glutathione. Here, a simple but very efficient assay was built up for screening reactive drug metabolites, utilizing stable isotope labeled glutathione, potassium cyanide and semicarbazide as trapping agents and highly sensitive ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC/TOFMS) as an analytical tool. A group of twelve structurally different compounds was used as a test set, and a large number of trapped metabolites were detected for most of them, including many conjugates not reported previously. Glutathione-trapped metabolites were detected for nine of the twelve test compounds, whereas cyanide-trapped metabolites were found for eight and semicarbazide-trapped for three test compounds. The high mass accuracy of TOFMS provided unambiguous identification of change in molecular formula by formation of a reactive metabolite. In addition, use of a mass defect filter was found to be a usable tool when mining the trapped conjugates from the acquired data. The approach was shown to provide superior detection sensitivity in comparison to traditional methods based on neutral loss or precursor ion scanning with a triple quadrupole mass spectrometer, and clearly more efficient detection and characterization of reactive drug metabolites with a simpler test setup.
Collapse
Affiliation(s)
- Timo Rousu
- Novamass Ltd., Medipolis Center, Kiviharjuntie 11, 90220 Oulu, Finland
| | | | | |
Collapse
|
25
|
Ott H, Bergström MA, Heise R, Skazik C, Zwadlo-Klarwasser G, Merk HF, Baron JM, Karlberg AT. Cutaneous Metabolic Activation of Carvoxime, a Self-Activating, Skin-Sensitizing Prohapten. Chem Res Toxicol 2009; 22:399-405. [DOI: 10.1021/tx8003642] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hagen Ott
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Moa Andresen Bergström
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Ruth Heise
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Claudia Skazik
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Gabriele Zwadlo-Klarwasser
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Hans F. Merk
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Jens M. Baron
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| | - Ann-Therese Karlberg
- Department of Dermatology and Allergology and Interdisciplinary Centre for Clinical Research (IZKF) BIOMAT, RWTH Aachen University, D-52074 Aachen, Germany, and Dermatochemistry and Skin Allergy, Department of Chemistry, Göteborg University, SE-412 96, Göteborg, Sweden
| |
Collapse
|
26
|
Tolonen A, Turpeinen M, Pelkonen O. Liquid chromatography-mass spectrometry in in vitro drug metabolite screening. Drug Discov Today 2008; 14:120-33. [PMID: 19059358 DOI: 10.1016/j.drudis.2008.11.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 10/15/2008] [Accepted: 11/05/2008] [Indexed: 12/31/2022]
Abstract
A combination of high performance liquid chromatography (HPLC) and mass spectrometry (LC/MS) has proven its status as the most powerful analytical tool for screening and identifying drug metabolites in modern drug discovery. These techniques have become irreplaceable for drug metabolism laboratories, providing high amounts of information from a wide variety of samples. This review focuses on the most common and useful applications of these techniques when working on in vitro metabolism, more specifically with screening and identification of chemically stable or reactive metabolites formed via biotransformation reactions. Matching specific tasks and suitable instruments is a recurring consideration; for many reasons, the time-of-flight or orbitrap mass spectrometry provides clearly increased efficiency in metabolite profiling compared to other types of mass spectrometry.
Collapse
Affiliation(s)
- Ari Tolonen
- Novamass Ltd., Medipolis Center, Kiviharjuntie 11, 90220 Oulu, Finland.
| | | | | |
Collapse
|
27
|
Kalapila AG, Loktionova NA, Pegg AE. Alkyltransferase-mediated toxicity of 1,3-butadiene diepoxide. Chem Res Toxicol 2008; 21:1851-61. [PMID: 18712882 DOI: 10.1021/tx800178t] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human O(6)-alkylguanine-DNA alkyltransferase (hAGT) expression increases mutations and cytotoxicity following exposure to 1,3-butadiene diepoxide (BDO), and hAGT-DNA cross-links are formed in the presence of BDO. We have used hAGT mutants to investigate the mechanism of cross-link formation and genotoxicity. Formation of a hAGT-DNA conjugate in vitro was observed with C145S and C145A mutant proteins but was considerably diminished with the C145A/C150S double mutant confirming that cross-linking primarily involves either of these two cysteine residues, which are located in the active site pocket of the protein. Cross-link formation by BDO occurred both via (a) an initial reaction of BDO with hAGT followed by attack of the reactive hAGT complex on DNA, and (b) the initial reaction of BDO with DNA followed by a reaction between hAGT and the DNA adduct. These results differ from those with 1,2-dibromoethane (DBE) where Cys(145) is the only site of attachment and pathway (b) does not occur. The complex formed between hAGT at Cys(145) and BDO was very unstable in aqueous solution. However, the BDO-hAGT complex at Cys(150) exhibited stability for more than 1 h. The effect of hAGT and mutants on BDO-induced genotoxicity was studied in E. coli using the forward assay to rifampicin resistance. Both mutations and cell killing were greatly increased by wild type hAGT, and there was a smaller but significant effect with the C145A mutant. The R128A mutant and R128A/C145A and C145A/C150S double mutants were ineffective, supporting the hypothesis that the formation of hAGT-DNA cross-links is responsible for the enhanced genotoxicity detected in this biological system. In the absence of hAGT, there were equal proportions of G:C to A:T transitions, G:C to T:A transversions, and A:T to T:A transversions. Wild type hAGT expression yielded significantly greater G:C to A:T and A:T to G:C transitions, whereas C145A mutant expression resulted in more transitions and transversions at A:T base-pairs.
Collapse
Affiliation(s)
- Aley G Kalapila
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | |
Collapse
|
28
|
Mutlib AE. Application of stable isotope-labeled compounds in metabolism and in metabolism-mediated toxicity studies. Chem Res Toxicol 2008; 21:1672-89. [PMID: 18702535 DOI: 10.1021/tx800139z] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stable isotope-labeled compounds have been synthesized and utilized by scientists from various areas of biomedical research during the last several decades. Compounds labeled with stable isotopes, such as deuterium and carbon-13, have been used effectively by drug metabolism scientists and toxicologists to gain better understanding of drugs' disposition and their potential role in target organ toxicities. The combination of stable isotope-labeling techniques with mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, which allows rapid acquisition and interpretation of data, has promoted greater use of these stable isotope-labeled compounds in absorption, distribution, metabolism, and excretion (ADME) studies. Examples of the use of stable isotope-labeled compounds in elucidating structures of metabolites and delineating complex metabolic pathways are presented in this review. The application of labeled compounds in mechanistic toxicity studies will be discussed by providing an example of how strategic placement of a deuterium atom in a drug molecule mitigated specific-specific renal toxicity. Other examples from the literature demonstrating the application of stable isotope-labeled compounds in understanding metabolism-mediated toxicities are presented. Furthermore, an example of how a stable isotope-labeled compound was utilized to better understand some of the gene changes in toxicogenomic studies is discussed. The interpretation of large sets of data produced from toxicogenomics studies can be a challenge. One approach that could be used to simplify interpretation of the data, especially from studies designed to link gene changes with the formation of reactive metabolites thought to be responsible for toxicities, is through the use of stable isotope-labeled compounds. This is a relatively unexplored territory and needs to be further investigated. The employment of analytical techniques, especially mass spectrometry and NMR, used in conjunction with stable isotope-labeled compounds to establish and understand mechanistic link between reactive metabolite formation, genomic, and proteomic changes and onset of toxicity is proposed. The use of stable isotope-labeled compounds in early human ADME studies as a way of identifying and possibly quantifying all drug-related components present in systemic circulation is suggested.
Collapse
Affiliation(s)
- Abdul E Mutlib
- Biotransformation Department, Drug Safety and Metabolism, Wyeth Research, Collegeville, Pennsylvania 19426, USA.
| |
Collapse
|
29
|
Gerberick F, Aleksic M, Basketter D, Casati S, Karlberg AT, Kern P, Kimber I, Lepoittevin JP, Natsch A, Ovigne JM, Rovida C, Sakaguchi H, Schultz T. Chemical reactivity measurement and the predicitve identification of skin sensitisers. The report and recommendations of ECVAM Workshop 64. Altern Lab Anim 2008; 36:215-42. [PMID: 18522487 DOI: 10.1177/026119290803600210] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Frank Gerberick
- Procter & Gamble Company, Miami Valley Innovation Center, Cincinnati, OH 45253, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Dereszynski DM, Center SA, Randolph JF, Brooks MB, Hadden AG, Palyada KS, McDonough SP, Messick J, Stokol T, Bischoff KL, Gluckman S, Sanders SY. Clinical and clinicopathologic features of dogs that consumed foodborne hepatotoxic aflatoxins: 72 cases (2005–2006). J Am Vet Med Assoc 2008; 232:1329-37. [DOI: 10.2460/javma.232.9.1329] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Mahajan MK, Evans CA. Dual negative precursor ion scan approach for rapid detection of glutathione conjugates using liquid chromatography/tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2008; 22:1032-1040. [PMID: 18320543 DOI: 10.1002/rcm.3458] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Screening for conjugates formed by the tripeptide glutathione with new chemical entities is an essential step during the drug discovery process, as the formation of these conjugates serves as an indicator for the presence of reactive electrophilic intermediates. To increase the selectivity and throughput of this analysis, various mass spectral scan types have evolved over time. Historically, samples were analyzed under positive ionization conditions for the neutral loss of m/z 129 (loss of the pyroglutamic acid moiety from glutathione); however, more recently, negative precursor ion scanning for the loss of m/z 272 (deprotonated gamma-glutamyl-dehydroalanyl-glycine from glutathione) has emerged as a more selective tool. Further increasing the selectivity, we report on an extension of this methodology by incorporating a simultaneous dual negative precursor ion scan for two commonly observed ion fragments from glutathione conjugates, m/z 272 and 254 (the dehydrated form of m/z 272). This negative dual precursor ion scan methodology was first validated using substrates known to undergo metabolic bioactivation (diclofenac, carbamazepine, and 3-methyl indole) and has then been applied to the routine analysis of proprietary compounds undergoing active lead optimization. In comparison to alternative scan methodologies, the increased selectivity offered by this simultaneous dual precursor method results in a reduction in the generation of false positive results as well as reduced data interpretation time.
Collapse
Affiliation(s)
- Mukesh K Mahajan
- Preclinical Drug Discovery, Cardiovascular and Urogenital Center of Excellence for Drug Discovery, GlaxoSmithKline, King of Prussia, PA 19406, USA.
| | | |
Collapse
|
32
|
Druckova A, Mernaugh RL, Ham AJL, Marnett LJ. Identification of the Protein Targets of the Reactive Metabolite of Teucrin A in Vivo in the Rat. Chem Res Toxicol 2007; 20:1393-408. [PMID: 17892266 DOI: 10.1021/tx7001405] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Covalent modification of proteins is associated with the toxicity of many electrophiles, and the identification of relevant in vivo protein targets is a desirable but challenging goal. Here, we describe a strategy for the enrichment of adducted proteins utilizing single-chain fragment variable (ScFv) antibodies selected using phage-display technology. Teucrin A is a furan-containing diterpenoid found in the herb germander that is primarily responsible for the herb's hepatotoxicity in rodents and humans following metabolic activation by cytochrome P450 enzymes. Conjugates of the 1,4-enedial derivative of teucrin A, its presumed toxic metabolite, with lysine- and cysteine-containing peptides were synthesized and used to select ScFvs from a rodent phage-displayed library, which recognized the terpenoid moiety of the teucrin-derived adducts. Immunoaffinity isolation of adducted proteins from rat liver homogenates following administration of a toxic dose of teucrin A afforded a family of proteins that were identified by liquid chromatography/tandem mass spectrometry. Of the 46 proteins identified in this study, most were of mitochondrial and endoplasmic reticulum origin. Several cytosolic proteins were found, as well as four peroxisomal and two secreted proteins. Using Ingenuity Pathway Analysis software, two significant networks involving the target genes were identified that had major functions in gene expression, small molecule biochemistry, and cellular function and maintenance. These included proteins involved in lipid, amino acid, and drug metabolism. This study illustrates the utility of chemically synthesized biological conjugates of reactive intermediates and the potential of the phage display technology for the generation of affinity reagents for the isolation of adducted proteins.
Collapse
Affiliation(s)
- Alexandra Druckova
- Department of Biochemistry, A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | | | | | | |
Collapse
|
33
|
Cardador-Martínez A, Albores A, Bah M, Calderón-Salinas V, Castaño-Tostado E, Guevara-González R, Shimada-Miyasaka A, Loarca-Piña G. Relationship among antimutagenic, antioxidant and enzymatic activities of methanolic extract from common beans (Phaseolus vulgaris L). PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2006; 61:161-8. [PMID: 17048099 DOI: 10.1007/s11130-006-0026-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Common beans are rich in phenolic compounds, which can provide health benefits to the consumer. The objective of this work was to study the relationship among antimutagenicity, antioxidant and enzymatic activities of methanolic extract and trolox by principal components multivariate analysis. Antimutagenicity of phenolic compounds present in methanolic extract from the seed coat of common beans (P. vulgaris, Flor de Mayo Bajío cultivar) and trolox against AFB1 mutagenicity were evaluated in the Salmonella typhimurium microsuspension assay. Antioxidant capacity of methanolic extract and trolox were evaluated using beta-carotene and 1,1-diphenyl-2-picryhydrazyl (DPPH) in vitro model assays. Cythrome P450 activity was measured by fluorometric assay. For phenolic extract, trolox and phenolic extract+trolox, the inhibition on AFB1 mutagenicity in tester strain TA100 was 47, 59 and 69%, respectively. While in TA98 was 39, 48 and 68%. The inhibition of phenolic compounds, trolox and phenolic compounds+trolox on cytochrome P450 (CYP450) activity was 48, 59 and 88%, respectively. Correlation analysis showed that phenolic extract and trolox have high antimutagenic and antioxidant activity and also inhibited enzymatic activity. The results suggest that the primary mechanism of action of phenolic compounds in beans against AFB1 mutagenicity may be extra-cellular in the microsuspension assay.
Collapse
Affiliation(s)
- Anaberta Cardador-Martínez
- PROPAC (Programa de Posgrado en Alimentos del Centro de la República), Research and Graduated Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Qro., 76010, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Loeber R, Rajesh M, Fang Q, Pegg AE, Tretyakova N. Cross-linking of the human DNA repair protein O6-alkylguanine DNA alkyltransferase to DNA in the presence of 1,2,3,4-diepoxybutane. Chem Res Toxicol 2006; 19:645-54. [PMID: 16696566 PMCID: PMC3213031 DOI: 10.1021/tx0600088] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1,2,3,4-Diepoxybutane (DEB) is a key carcinogenic metabolite of the important industrial chemical 1,3-butadiene. DEB is a bifunctional alkylating agent capable of reacting with DNA and proteins. Initial DNA alkylation by DEB produces N7-(2'-hydroxy-3',4'-epoxybut-1'-yl)-guanine monoadducts, which can react with another nucleophilic site to form cross-linked adducts. A recent report revealed a strong correlation between cellular expression of the DNA repair protein O6-alkylguanine DNA alkyltransferase (AGT) and the cytotoxic and mutagenic activity of DEB, suggesting that DEB induces AGT-DNA cross-links (Valadez, J. G., et al. (2004) Activation of bis-electrophiles to mutagenic conjugates by human O6-alkylguanine-DNA alkyltransferase. Chem. Res. Toxicol. 17, 972-982). The purpose of our study was to analyze the formation and structures of DEB-induced AGT-DNA conjugates and to identify specific amino acid residues within the protein involved in cross-linking. DNA-protein cross-link formation was detected by SDS-PAGE when 32P-labeled double-stranded oligodeoxynucleotides were exposed to DEB in the presence of either wild-type hAGT or a C145A hAGT mutant. Capillary HPLC-electrospray ionization mass spectrometry (ESI-MS) analysis of hAGT that had been treated with N7-(2'-hydroxy-3',4'-epoxybut-1'-yl)-deoxyguanosine (dG monoepoxide) revealed the ability of the protein to form either one or two butanediol-dG cross-links, corresponding to mass shifts of +353 and +706 Da, respectively. HPLC-ESI+ -MS/MS sequencing of the tryptic peptides obtained from dG monoepoxide-treated protein indicated that the two cross-linking sites were the alkyl acceptor site, Cys145, and a neighboring active site residue, Cys150. The same two amino acid residues of hAGT became covalently cross-linked to DNA following DEB treatment. Modification of Cys145 was further confirmed by HPLC-ESI+ -MS/MS analysis of dG monoepoxide-treated synthetic peptide GNPVPILIPCHR which represents the active site tryptic fragment of hAGT (C = Cys145). The replacement of the catalytic cysteine residue with alanine in the C145A hAGT mutant abolished DEB-induced cross-linking at this site, while the formation of conjugates via neighboring Cys150 was retained. The exact chemical structure of the cross-linked lesion was established as 1-(S-cysteinyl)-4-(guan-7-yl)-2,3-butanediol by HPLC-ESI+ -MS/MS analysis of the amino acids resulting from the total digestion of modified proteins analyzed in parallel with an authentic standard. AGT-DNA cross-linking is a likely mechanism of DEB-mediated cytotoxicity in cells expressing this important repair protein.
Collapse
Affiliation(s)
| | | | - Qingming Fang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Anthony E. Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Natalia Tretyakova
- Corresponding author: University of Minnesota Cancer Center, Mayo Mail Code 806, Room 760 E CCRB, 420 Delaware St SE, Minneapolis, MN 55455, USA; tel: (612) 626-3432 fax: (612) 626-5135
| |
Collapse
|
35
|
Affiliation(s)
- Patrick J O'Brien
- Department of Biological Chemistry, University of Michigan, Ann Arbor, 48109-0606, USA.
| |
Collapse
|
36
|
Abstract
The cytochrome P450 (P450) enzymes are the major catalysts involved in the metabolism of drugs. Bioavailability and toxicity are 2 of the most common barriers in drug development today, and P450 and the conjugation enzymes can influence these effects. The toxicity of drugs can be considered in 5 contexts: on-target toxicity, hypersensitivity and immunological reactions, off-target pharmacology, bioactivation to reactive intermediates, and idiosyncratic drug reactions. The chemistry of bioactivation is reasonably well understood, but the mechanisms underlying biological responses are not. In the article we consider what fraction of drug toxicity actually involves metabolism, and we examine how species and human interindividual variations affect pharmacokinetics and toxicity.
Collapse
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
37
|
Furge LL, Guengerich FP. Cytochrome P450 enzymes in drug metabolism and chemical toxicology: An introduction. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2006; 34:66-74. [PMID: 21638641 DOI: 10.1002/bmb.2006.49403402066] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Cytochrome P450 (P450) enzymes include a family of related enzymes that are involved in metabolism of vitamins, steroids, fatty acids, and other chemicals. This review presents a brief historical overview of the discovery and characterization of P450 enzymes extending from intermediary metabolism to the fields of drug metabolism and toxicology. Introductions to P450 enzyme structure and function are also presented. The goals of this review are to 1) provide an introduction to a few of the many aspects of P450 research relating to humans, 2) introduce additional ways of thinking about metabolism, 3) provide some basic examples of P450 enzymology, and 4) provide applications to topics widely taught in undergraduate courses in biochemistry.
Collapse
Affiliation(s)
- Laura Lowe Furge
- Department of Chemistry, Kalamazoo College, Kalamazoo, Michigan 49006-3295.
| | | |
Collapse
|
38
|
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA.
| |
Collapse
|