1
|
Georgiou-Siafis SK, Tsiftsoglou AS. The Key Role of GSH in Keeping the Redox Balance in Mammalian Cells: Mechanisms and Significance of GSH in Detoxification via Formation of Conjugates. Antioxidants (Basel) 2023; 12:1953. [PMID: 38001806 PMCID: PMC10669396 DOI: 10.3390/antiox12111953] [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: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione (GSH) is a ubiquitous tripeptide that is biosynthesized in situ at high concentrations (1-5 mM) and involved in the regulation of cellular homeostasis via multiple mechanisms. The main known action of GSH is its antioxidant capacity, which aids in maintaining the redox cycle of cells. To this end, GSH peroxidases contribute to the scavenging of various forms of ROS and RNS. A generally underestimated mechanism of action of GSH is its direct nucleophilic interaction with electrophilic compounds yielding thioether GSH S-conjugates. Many compounds, including xenobiotics (such as NAPQI, simvastatin, cisplatin, and barbital) and intrinsic compounds (such as menadione, leukotrienes, prostaglandins, and dopamine), form covalent adducts with GSH leading mainly to their detoxification. In the present article, we wish to present the key role and significance of GSH in cellular redox biology. This includes an update on the formation of GSH-S conjugates or GSH adducts with emphasis given to the mechanism of reaction, the dependence on GST (GSH S-transferase), where this conjugation occurs in tissues, and its significance. The uncovering of the GSH adducts' formation enhances our knowledge of the human metabolome. GSH-hematin adducts were recently shown to have been formed spontaneously in multiples isomers at hemolysates, leading to structural destabilization of the endogenous toxin, hematin (free heme), which is derived from the released hemoglobin. Moreover, hemin (the form of oxidized heme) has been found to act through the Kelch-like ECH associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor-2 (Nrf2) signaling pathway as an epigenetic modulator of GSH metabolism. Last but not least, the implications of the genetic defects in GSH metabolism, recorded in hemolytic syndromes, cancer and other pathologies, are presented and discussed under the framework of conceptualizing that GSH S-conjugates could be regarded as signatures of the cellular metabolism in the diseased state.
Collapse
Affiliation(s)
| | - Asterios S. Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
| |
Collapse
|
2
|
Kimura Y, Ekuban FA, Zong C, Sugie S, Zhang X, Itoh K, Yamamoto M, Ichihara S, Ohsako S, Ichihara G. Role of Nrf2 in 1,2-dichloropropane-induced cell proliferation and DNA damage in the mouse liver. Toxicol Sci 2023; 195:28-41. [PMID: 37326970 DOI: 10.1093/toxsci/kfad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
1,2-Dichloropropane (1,2-DCP) is recognized as the causative chemical of occupational cholangiocarcinoma in printing workers in Japan. However, the cellular and molecular mechanisms of 1,2-DCP-induced carcinogenesis remains elusive. The present study investigated cellular proliferation, DNA damage, apoptosis, and expression of antioxidant and proinflammatory genes in the liver of mice exposed daily to 1,2-DCP for 5 weeks, and the role of nuclear factor erythroid 2-related factor 2 (Nrf2) in these responses. Wild-type and Nrf2-knockout (Nrf2-/-) mice were administered 1,2-DCP by gastric gavage, and then the livers were collected for analysis. Immunohistochemistry for BrdU or Ki67 and TUNEL assay revealed that exposure to 1,2-DCP dose-dependently increased proliferative cholangiocytes, whereas decreased apoptotic cholangiocytes in wild-type mice but not in Nrf2-/- mice. Western blot and quantitative real-time PCR showed that exposure to 1,2-DCP increased the levels of DNA double-strand break marker γ-H2AX and mRNA expression levels of NQO1, xCT, GSTM1, and G6PD in the livers of wild-type mice in a dose-dependent manner, but no such changes were noted in Nrf2-/- mice. 1,2-DCP increased glutathione levels in the liver of both the wild-type and Nrf2-/- mice, suggesting that an Nrf2-independent mechanism contributes to 1,2-DCP-induced increase in glutathione level. In conclusion, the study demonstrated that exposure to 1,2-DCP induced proliferation but reduced apoptosis in cholangiocytes, and induced double-strand DNA breaks and upregulation of antioxidant genes in the liver in an Nrf2-dependent manner. The study suggests a role of Nrf2 in 1,2-DCP-induced cell proliferation, antiapoptotic effect, and DNA damage, which are recognized as key characteristics of carcinogens.
Collapse
Affiliation(s)
- Yusuke Kimura
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan
| | - Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan
| | - Shigeyuki Sugie
- Department of Diagnostic Pathology, Asahi University Murakami Memorial Hospital, Gifu 550-8856, Japan
| | - Xiao Zhang
- Department of Toxicology, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou 510300, People's Republic of China
| | - Ken Itoh
- Department of Stress Response Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Masayuki Yamamoto
- Division of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0431, Japan
| | - Seiichiro Ohsako
- Department of Environmental and Preventive Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan
| |
Collapse
|
3
|
Mechanistic and kinetic insights into the atmospheric degradation of (CH3)3CF and (CH3)3CCl initiated by Cl atom. COMPUT THEOR CHEM 2022. [DOI: 10.1016/j.comptc.2022.113807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
4
|
Ishida K, Werner JA, Davies R, Fan F, Thomas B, Wahlstrom J, Lipford JR, Monticello T. Nonclinical Safety Profile of Sotorasib, a KRAS G12C-Specific Covalent Inhibitor for the Treatment of KRAS p.G12C-Mutated Cancer. Int J Toxicol 2021; 40:427-441. [PMID: 34137282 DOI: 10.1177/10915818211022965] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sotorasib is a first-in-class KRASG12C covalent inhibitor in clinical development for the treatment of tumors with the KRAS p.G12C mutation. A comprehensive nonclinical safety assessment package, including secondary/safety pharmacology and toxicology studies, was conducted to support the marketing application for sotorasib. Sotorasib was negative in a battery of genotoxicity assays and negative in an in vitro phototoxicity assay. Based on in vitro assays, sotorasib had no off-target effects against various receptors, enzymes (including numerous kinases), ion channels, or transporters. Consistent with the tumor-specific target distribution (ie, KRASG12C), there were no primary pharmacology-related on-target effects identified. The kidney was identified as a target organ in the rat but not the dog. Renal toxicity in the rat was characterized by tubular degeneration and necrosis restricted to a specific region suggesting that the toxicity was attributed to the local formation of a putative toxic reactive metabolite. In the 3-month dog study, adaptive changes of hepatocellular hypertrophy due to drug metabolizing enzyme induction were observed in the liver that was associated with secondary effects in the pituitary and thyroid gland. Sotorasib was not teratogenic and had no direct effect on embryo-fetal development in the rat or rabbit. Human, dog, and rat circulating metabolites, M24, M10, and M18, raised no clinically relevant safety concerns based on the general toxicology studies, primary/secondary pharmacology screening, an in vitro human ether-à-go-go-related gene assay, or mutagenicity assessment. Overall, the results of the nonclinical safety program support a high benefit/risk ratio of sotorasib for the treatment of patients with KRAS p.G12C-mutated tumors.
Collapse
Affiliation(s)
| | | | | | - Fan Fan
- Amgen Inc, Research, Thousand Oaks, CA, USA
| | | | | | | | | |
Collapse
|
5
|
Jain S, Norinder U, Escher SE, Zdrazil B. Combining In Vivo Data with In Silico Predictions for Modeling Hepatic Steatosis by Using Stratified Bagging and Conformal Prediction. Chem Res Toxicol 2020; 34:656-668. [PMID: 33347274 PMCID: PMC7887803 DOI: 10.1021/acs.chemrestox.0c00511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatic steatosis (fatty liver) is a severe liver disease induced by the excessive accumulation of fatty acids in hepatocytes. In this study, we developed reliable in silico models for predicting hepatic steatosis on the basis of an in vivo data set of 1041 compounds measured in rodent studies with repeated oral exposure. The imbalanced nature of the data set (1:8, with the "steatotic" compounds belonging to the minority class) required the use of meta-classifiers-bagging with stratified under-sampling and Mondrian conformal prediction-on top of the base classifier random forest. One major goal was the investigation of the influence of different descriptor combinations on model performance (tested by predicting an external validation set): physicochemical descriptors (RDKit), ToxPrint features, as well as predictions from in silico nuclear receptor and transporter models. All models based upon descriptor combinations including physicochemical features led to reasonable balanced accuracies (BAs between 0.65 and 0.69 for the respective models). Combining physicochemical features with transporter predictions and further with ToxPrint features gave the best performing model (BAs up to 0.7 and efficiencies of 0.82). Whereas both meta-classifiers proved useful for this highly imbalanced toxicity data set, the conformal prediction framework also guarantees the error level and thus might be favored for future studies in the field of predictive toxicology.
Collapse
Affiliation(s)
- Sankalp Jain
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Ulf Norinder
- Unit of Toxicology Sciences, Swetox, Karolinska Institutet, SE-15136 Södertälje, Sweden
| | - Sylvia E Escher
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), 30625 Hannover, Germany
| | - Barbara Zdrazil
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Abaukaka YA, Sanusi S, Ozigi KA, Malo FU. Assessment of the cytotoxic and mutagenic potential of dichlorvos (DDVP) using in silico classification model; a health hazard awareness in Nigeria. Environ Anal Health Toxicol 2020; 35:e2020016. [PMID: 32979901 PMCID: PMC7656162 DOI: 10.5620/eaht.2020016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 11/11/2022] Open
Abstract
Dichlorvos (DDVP) has been abused in Nigeria for suicide attempts, topical applications to treat an ectoparasitic infestation, and indiscriminate use on farm produce. Exposure to this compound in subacute concentration can cause toxicity in different tissues by alteration of the cellular antioxidative defence mechanism. This analysis is aimed at the systematic profiling of DDVP to assess its cytotoxic and mutagenic potential for human vulnerability using an in silico classification model. DDVP was grouped into categories of analogue chemical compounds generated from inventories based on structural alerts that measure the biological effects on cell lines and animal models using the quantitative structure-activity relationship (QSAR) model. The cytotoxic and mutagenic potential of DDVP was assessed by analyzing target endpoints like skin sensitization, oral/inhalation toxicity, neurotoxicity and mutagenicity. DDVP shows moderate sensitization potential that can induce skin irritation during prolonged exposure because of the presence of dichlorovenyl side-chain that interacts with cellular proteins and causes degradation. 50% lethal dose (LD50) of DDVP per body weight was determined to be 26.2 mg/kg in a rat model at 95% confidence range for acute oral toxicity, and 14.4 mmol/L was estimated as 50% lethal concentration (LC50) in the atmosphere due to acute inhalation toxicity. DDVP can also inhibit acetylcholinesterase in the nervous system to produce nicotinic and muscarinic symptoms like nausea, vomiting, lacrimation, salivation, bradycardia, and respiratory failure may cause death. The widely used pesticide causes weak DNA methylation which can repress gene transcription on promoter sites. DDVP is volatile so it can cause oral and inhalation toxicity coupled with neurotoxicity during prolonged exposure. Serum cholinesterase blood tests should be encouraged in federal and state hospitals to investigate related health challenges as DDVP is still used in Nigeria.
Collapse
Affiliation(s)
| | - Salihu Sanusi
- Tehran University of Medical Sciences, No. 226, Qods St., Keshavarz Blvd., Tehran, Iran
| | - Kabir Abdullahi Ozigi
- Tehran University of Medical Sciences, No. 226, Qods St., Keshavarz Blvd., Tehran, Iran
| | | |
Collapse
|
7
|
Coecke S, Ahr H, Blaauboer BJ, Bremer S, Casati S, Castell J, Combes R, Corvi R, Crespi CL, Cunningham ML, Elaut G, Eletti B, Freidig A, Gennari A, Ghersi-Egea JF, Guillouzo A, Hartung T, Hoet P, Ingelman-Sundberg M, Munn S, Janssens W, Ladstetter B, Leahy D, Long A, Meneguz A, Monshouwer M, Morath S, Nagelkerke F, Pelkonen O, Ponti J, Prieto P, Richert L, Sabbioni E, Schaack B, Steiling W, Testai E, Vericat JA, Worth A. Metabolism: A Bottleneck in In Vitro Toxicological Test Development. Altern Lab Anim 2019; 34:49-84. [PMID: 16522150 DOI: 10.1177/026119290603400113] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sandra Coecke
- ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
Many potentially toxic electrophilic xenobiotics and some endogenous compounds are detoxified by conversion to the corresponding glutathione S-conjugate, which is metabolized to the N-acetylcysteine S-conjugate (mercapturate) and excreted. Some mercapturate pathway components, however, are toxic. Bioactivation (toxification) may occur when the glutathione S-conjugate (or mercapturate) is converted to a cysteine S-conjugate that undergoes a β-lyase reaction. If the sulfhydryl-containing fragment produced in this reaction is reactive, toxicity may ensue. Some drugs and halogenated workplace/environmental contaminants are bioactivated by this mechanism. On the other hand, cysteine S-conjugate β-lyases occur in nature as a means of generating some biologically useful sulfhydryl-containing compounds.
Collapse
|
9
|
De La Rosa VY, Asfaha J, Fasullo M, Loguinov A, Li P, Moore LE, Rothman N, Nakamura J, Swenberg JA, Scelo G, Zhang L, Smith MT, Vulpe CD. Editor's Highlight: High-Throughput Functional Genomics Identifies Modulators of TCE Metabolite Genotoxicity and Candidate Susceptibility Genes. Toxicol Sci 2017; 160:111-120. [PMID: 28973557 PMCID: PMC5837773 DOI: 10.1093/toxsci/kfx159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Trichloroethylene (TCE), an industrial chemical and environmental contaminant, is a human carcinogen. Reactive metabolites are implicated in renal carcinogenesis associated with TCE exposure, yet the toxicity mechanisms of these metabolites and their contribution to cancer and other adverse effects remain unclear. We employed an integrated functional genomics approach that combined functional profiling studies in yeast and avian DT40 cell models to provide new insights into the specific mechanisms contributing to toxicity associated with TCE metabolites. Genome-wide profiling studies in yeast identified the error-prone translesion synthesis (TLS) pathway as an import mechanism in response to TCE metabolites. The role of TLS DNA repair was further confirmed by functional profiling in DT40 avian cell lines, but also revealed that TLS and homologous recombination DNA repair likely play competing roles in cellular susceptibility to TCE metabolites in higher eukaryotes. These DNA repair pathways are highly conserved between yeast, DT40, and humans. We propose that in humans, mutagenic TLS is favored over homologous recombination repair in response to TCE metabolites. The results of these studies contribute to the body of evidence supporting a mutagenic mode of action for TCE-induced renal carcinogenesis mediated by reactive metabolites in humans. Our approach illustrates the potential for high-throughput in vitro functional profiling in yeast to elucidate toxicity pathways (molecular initiating events, key events) and candidate susceptibility genes for focused study.
Collapse
Affiliation(s)
- Vanessa Y. De La Rosa
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720
| | - Jonathan Asfaha
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720
| | - Michael Fasullo
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, New York 12205
| | - Alex Loguinov
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720
| | - Peng Li
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Lee E. Moore
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jun Nakamura
- Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | | | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, California 94720
| | - Martyn T. Smith
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, California 94720
| | - Chris D. Vulpe
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720
| |
Collapse
|
10
|
Three common pathways of nephrotoxicity induced by halogenated alkenes. Cell Biol Toxicol 2015; 31:1-13. [DOI: 10.1007/s10565-015-9293-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/29/2015] [Indexed: 12/13/2022]
|
11
|
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: 1.4] [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
|
12
|
Pinto JT, Krasnikov BF, Alcutt S, Jones ME, Dorai T, Villar MT, Artigues A, Li J, Cooper AJL. Kynurenine aminotransferase III and glutamine transaminase L are identical enzymes that have cysteine S-conjugate β-lyase activity and can transaminate L-selenomethionine. J Biol Chem 2014; 289:30950-61. [PMID: 25231977 DOI: 10.1074/jbc.m114.591461] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Three of the four kynurenine aminotransferases (KAT I, II, and IV) that synthesize kynurenic acid, a neuromodulator, are identical to glutamine transaminase K (GTK), α-aminoadipate aminotransferase, and mitochondrial aspartate aminotransferase, respectively. GTK/KAT I and aspartate aminotransferase/KAT IV possess cysteine S-conjugate β-lyase activity. The gene for the former enzyme, GTK/KAT I, is listed in mammalian genome data banks as CCBL1 (cysteine conjugate beta-lyase 1). Also listed, despite the fact that no β-lyase activity has been assigned to the encoded protein in the genome data bank, is a CCBL2 (synonym KAT III). We show that human KAT III/CCBL2 possesses cysteine S-conjugate β-lyase activity, as does mouse KAT II. Thus, depending on the nature of the substrate, all four KATs possess cysteine S-conjugate β-lyase activity. These present studies show that KAT III and glutamine transaminase L are identical enzymes. This report also shows that KAT I, II, and III differ in their ability to transaminate methyl-L-selenocysteine (MSC) and L-selenomethionine (SM) to β-methylselenopyruvate (MSP) and α-ketomethylselenobutyrate, respectively. Previous studies have identified these seleno-α-keto acids as potent histone deacetylase inhibitors. Methylselenol (CH3SeH), also purported to have chemopreventive properties, is the γ-elimination product of SM and the β-elimination product of MSC catalyzed by cystathionine γ-lyase (γ-cystathionase). KAT I, II, and III, in part, can catalyze β-elimination reactions with MSC generating CH3SeH. Thus, the anticancer efficacy of MSC and SM will depend, in part, on the endogenous expression of various KAT enzymes and cystathionine γ-lyase present in target tissue coupled with the ability of cells to synthesize in situ either CH3SeH and/or seleno-keto acid metabolites.
Collapse
Affiliation(s)
- John T Pinto
- From the Departments of Biochemistry and Molecular Biology and
| | | | - Steven Alcutt
- From the Departments of Biochemistry and Molecular Biology and
| | - Melanie E Jones
- From the Departments of Biochemistry and Molecular Biology and
| | - Thambi Dorai
- Urology, New York Medical College, Valhalla, New York 10595
| | - Maria T Villar
- the Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, and
| | - Antonio Artigues
- the Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, and
| | - Jianyong Li
- the Department of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061
| | | |
Collapse
|
13
|
Suzuki T, Yanagiba Y, Suda M, Wang RS. Assessment of the genotoxicity of 1,2-dichloropropane and dichloromethane after individual and co-exposure by inhalation in mice. J Occup Health 2014; 56:205-14. [PMID: 24739373 DOI: 10.1539/joh.13-0236-oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Occurrence of cholangiocarcinoma was recently reported at a high incidence rate among the employees working for an offset printing company in Osaka, Japan. 1,2-Dichloropropane (1,2-DCP) and dichloromethane (DCM) are suspected to be the causes of the cancer, as they had been used as ink cleaners in large amounts. However, it is not clear whether these chlorinated organic solvents played a role in the occurrence of cholangiocarcinoma or why the incidence rate is so high among the workers in this industry. To provide possible evidence for this severe occupational problem, we investigated the genotoxic effects of 1,2-DCP and DCM. METHODS Male B6C3F1 and gpt Delta C57BL/6J mice were exposed by inhalation to the individual solvents or both solvents at multiple concentrations including the levels that were possibly present in the workplaces. The genotoxicity was analyzed by Pig-a gene mutation and micronuclei assays in peripheral blood and gpt mutation and comet assays in the livers of mice after repeated inhalation of 1,2-DCP or/and DCM. RESULTS The Pig-a mutant frequencies and micronuclei incidences were not significantly increased by exposure of either 1,2-DCP or/and DCM at any concentration, suggesting there was no genotoxic potential in bone marrow for both solvents. In the liver, DNA damage, as measured by the comet assay, was dose dependently increased by 1,2-DCP but not by DCM. The gpt mutant frequency was 2.6-fold that of the controls in the co-exposure group. CONCLUSIONS These results indicate that 1,2-DCP showed stronger genotoxicity in the liver and that the genotoxic effects were greatly enhanced by simultaneous exposure to DCM.
Collapse
Affiliation(s)
- Tetsuya Suzuki
- Division of Health Effects Research, National Institute of Occupational Safety and Health
| | | | | | | |
Collapse
|
14
|
Lock EA, Zhang J, Checkoway H. Solvents and Parkinson disease: a systematic review of toxicological and epidemiological evidence. Toxicol Appl Pharmacol 2013; 266:345-55. [PMID: 23220449 PMCID: PMC3621032 DOI: 10.1016/j.taap.2012.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 11/12/2012] [Accepted: 11/14/2012] [Indexed: 12/25/2022]
Abstract
Parkinson disease (PD) is a debilitating neurodegenerative motor disorder, with its motor symptoms largely attributable to loss of dopaminergic neurons in the substantia nigra. The causes of PD remain poorly understood, although environmental toxicants may play etiologic roles. Solvents are widespread neurotoxicants present in the workplace and ambient environment. Case reports of parkinsonism, including PD, have been associated with exposures to various solvents, most notably trichloroethylene (TCE). Animal toxicology studies have been conducted on various organic solvents, with some, including TCE, demonstrating potential for inducing nigral system damage. However, a confirmed animal model of solvent-induced PD has not been developed. Numerous epidemiologic studies have investigated potential links between solvents and PD, yielding mostly null or weak associations. An exception is a recent study of twins indicating possible etiologic relations with TCE and other chlorinated solvents, although findings were based on small numbers, and dose-response gradients were not observed. At present, there is no consistent evidence from either the toxicological or epidemiologic perspective that any specific solvent or class of solvents is a cause of PD. Future toxicological research that addresses mechanisms of nigral damage from TCE and its metabolites, with exposure routes and doses relevant to human exposures, is recommended. Improvements in epidemiologic research, especially with regard to quantitative characterization of long-term exposures to specific solvents, are needed to advance scientific knowledge on this topic.
Collapse
Affiliation(s)
- Edward A Lock
- Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences, Byrom Street, Liverpool, UK.
| | | | | |
Collapse
|
15
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
16
|
Abstract
Considerable support exists for the roles of metabolism in modulating the carcinogenic properties of chemicals. In particular, many of these compounds are pro-carcinogens that require activation to electrophilic forms to exert genotoxic effects. We systematically analyzed the existing literature on the metabolism of carcinogens by human enzymes, which has been developed largely in the past 25 years. The metabolism and especially bioactivation of carcinogens are dominated by cytochrome P450 enzymes (66% of bioactivations). Within this group, six P450s--1A1, 1A2, 1B1, 2A6, 2E1, and 3A4--accounted for 77% of the P450 activation reactions. The roles of these P450s can be compared with those estimated for drug metabolism and should be considered in issues involving enzyme induction, chemoprevention, molecular epidemiology, interindividual variations, and risk assessment.
Collapse
|
17
|
Cooper AJL, Krasnikov BF, Pinto JT, Bruschi SA. Measurement of cysteine S-conjugate β-lyase activity. CURRENT PROTOCOLS IN TOXICOLOGY 2011; Chapter 4:Unit 4.36. [PMID: 20949433 DOI: 10.1002/0471140856.tx0436s44] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cysteine S-conjugate β-lyases are pyridoxal 5'-phosphate (PLP)-containing enzymes that catalyze the conversion of cysteine S-conjugates [RSCH(2)CH(NH(3) (+))CO(2) (-)] and selenium Se-conjugates [RSeCH(2)CH(NH(3) (+))CO(2) (-)] that contain a leaving group in the β position to pyruvate, ammonium and a sulfur-containing fragment (RSH) or selenium-containing fragment (RSeH), respectively. In mammals, at least ten PLP enzymes catalyze β-elimination reactions with such cysteine S-conjugates. All are enzymes involved in amino acid metabolism that do not normally catalyze a β-lyase reaction, but catalyze a non-physiological β-lyase side-reaction that depends on the electron-withdrawing properties of the -SR or -SeR moiety. In the case of cysteine S-conjugates, if the eliminated RSH is stable, the compound may be S-thiomethylated and excreted (thiomethyl shunt) or S-glucuronidated and harmlessly excreted. However, if RSH is chemically reactive, the cysteine S-conjugate may be toxic as a result of the β-lyase reaction. The cysteine S-conjugate β-lyase pathway is of particular interest to toxicologists because it is involved in the bioactivation (toxification) of halogenated alkenes and certain drugs. This unit provides protocols for the analysis of cysteine S-conjugate β-lyase activity.
Collapse
|
18
|
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: 1.4] [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
|
19
|
Mattes TE, Alexander AK, Coleman NV. Aerobic biodegradation of the chloroethenes: pathways, enzymes, ecology, and evolution. FEMS Microbiol Rev 2010; 34:445-75. [DOI: 10.1111/j.1574-6976.2010.00210.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
20
|
Cysteine S-conjugate β-lyases: important roles in the metabolism of naturally occurring sulfur and selenium-containing compounds, xenobiotics and anticancer agents. Amino Acids 2010; 41:7-27. [PMID: 20306345 DOI: 10.1007/s00726-010-0552-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 03/01/2010] [Indexed: 12/13/2022]
Abstract
Cysteine S-conjugate β-lyases are pyridoxal 5'-phosphate-containing enzymes that catalyze β-elimination reactions with cysteine S-conjugates that possess a good leaving group in the β-position. The end products are aminoacrylate and a sulfur-containing fragment. The aminoacrylate tautomerizes and hydrolyzes to pyruvate and ammonia. The mammalian cysteine S-conjugate β-lyases thus far identified are enzymes involved in amino acid metabolism that catalyze β-lyase reactions as non-physiological side reactions. Most are aminotransferases. In some cases the lyase is inactivated by reaction products. The cysteine S-conjugate β-lyases are of much interest to toxicologists because they play an important key role in the bioactivation (toxication) of halogenated alkenes, some of which are produced on an industrial scale and are environmental contaminants. The cysteine S-conjugate β-lyases have been reviewed in this journal previously (Cooper and Pinto in Amino Acids 30:1-15, 2006). Here, we focus on more recent findings regarding: (1) the identification of enzymes associated with high-M(r) cysteine S-conjugate β-lyases in the cytosolic and mitochondrial fractions of rat liver and kidney; (2) the mechanism of syncatalytic inactivation of rat liver mitochondrial aspartate aminotransferase by the nephrotoxic β-lyase substrate S-(1,1,2,2-tetrafluoroethyl)-L-cysteine (the cysteine S-conjugate of tetrafluoroethylene); (3) toxicant channeling of reactive fragments from the active site of mitochondrial aspartate aminotransferase to susceptible proteins in the mitochondria; (4) the involvement of cysteine S-conjugate β-lyases in the metabolism/bioactivation of drugs and natural products; and (5) the role of cysteine S-conjugate β-lyases in the metabolism of selenocysteine Se-conjugates. This review emphasizes the fact that the cysteine S-conjugate β-lyases are biologically more important than hitherto appreciated.
Collapse
|
21
|
Bateman TJ, Debenham JS, Madsen-Duggan C, Toupence RB, Walsh TF, Truong Q, Bradley SA, Doss GA, Kumar S, Reddy VBG. Glutathione S-transferase catalyzed desulfonylation of a sulfonylfuropyridine. Drug Metab Dispos 2010; 38:108-14. [PMID: 19797605 DOI: 10.1124/dmd.109.029801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
MRL-1, a cannabinoid receptor-1 inverse agonist, was a member of a lead candidate series for the treatment of obesity. In rats, MRL-1 is eliminated mainly via metabolism, followed by excretion of the metabolites into bile. The major metabolite M1, a glutathione conjugate of MRL-1, was isolated and characterized by liquid chromatography/mass spectrometry and NMR spectroscopic methods. The data suggest that the t-butylsulfonyl group at C-2 of furopyridine was displaced by the glutathionyl group. In vitro experiments using rat and monkey liver microsomes in the presence of reduced glutathione (GSH) showed that the formation of M1 was independent of NADPH and molecular oxygen, suggesting that this reaction was not mediated by an oxidative reaction and a glutathione S-transferase (GST) was likely involved in catalyzing this reaction. Furthermore, a rat hepatic GST was capable of catalyzing the conversion of MRL-1 to M1 in the presence of GSH. When a close analog of MRL-1, a p-chlorobenzenesulfonyl furopyridine derivative (MRL-2), was incubated with rat liver microsomes in the presence of GSH, p-chlorobenzene sulfinic acid (M2) was also identified as a product in addition to the expected M1. Based on these data, a mechanism is proposed involving direct nucleophilic addition of GSH to sulfonylfuropyridine, resulting in an unstable adduct that spontaneously decomposes to form M1 and M2.
Collapse
Affiliation(s)
- Thomas J Bateman
- Department of Drug Metabolism and Pharmacokinetics, Merck Research Laboratories, Rahway, New Jersey, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Many potentially toxic electrophiles react with glutathione to form glutathione S-conjugates in reactions catalyzed or enhanced by glutathione S-transferases. The glutathione S-conjugate is sequentially converted to the cysteinylglycine-, cysteine- and N-acetyl-cysteine S-conjugate (mercapturate). The mercapturate is generally more polar and water soluble than the parent electrophile and is readily excreted. Excretion of the mercapturate represents a detoxication mechanism. Some endogenous compounds, such as leukotrienes, prostaglandin (PG) A2, 15-deoxy-Δ12,14-PGJ2, and hydroxynonenal can also be metabolized to mercapturates and excreted. On occasion, however, formation of glutathione S- and cysteine S-conjugates are bioactivation events as the metabolites are mutagenic and/or cytotoxic. When the cysteine S-conjugate contains a strong electron-withdrawing group attached at the sulfur, it may be converted by cysteine S-conjugate β-lyases to pyruvate, ammonium and the original electrophile modified to contain an –SH group. If this modified electrophile is highly reactive then the enzymes of the mercapturate pathway together with the cysteine S-conjugate β-lyases constitute a bioactivation pathway. Some endogenous halogenated environmental contaminants and drugs are bioactivated by this mechanism. Recent studies suggest that coupling of enzymes of the mercapturate pathway to cysteine S-conjugate β-lyases may be more common in nature and more widespread in the metabolism of electrophilic xenobiotics than previously realized.
Collapse
|
23
|
Zhu X, Gallogly MM, Mieyal JJ, Anderson VE, Sayre LM. Covalent cross-linking of glutathione and carnosine to proteins by 4-oxo-2-nonenal. Chem Res Toxicol 2009; 22:1050-9. [PMID: 19480392 DOI: 10.1021/tx9000144] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The lipid oxidation product 4-oxo-2-nonenal (ONE) derived from peroxidation of polyunsaturated fatty acids is a highly reactive protein cross-linking reagent. The major family of cross-links reflects conjugate addition of side chain nucleophiles such as sulfhydryl or imidazole groups to the C triple bond C of ONE to give either a 2- or 3-substituted 4-ketoaldehyde, which then undergoes Paal-Knorr condensation with the primary amine of protein lysine side chains. If ONE is intercepted in biological fluids by antielectrophiles such as glutathione (GSH) or beta-alanylhistidine (carnosine), this would lead to circulating 4-ketoaldehydes that could then bind covalently to the protein Lys residues. This phenomenon was investigated by SDS-PAGE and mass spectrometry (matrix-assisted laser desorption/ionization time-of-flight and LC-ESI-MS/MS with both tryptic and chymotryptic digestion). Under the reaction conditions of 0.25-2 mM ONE, 1 mM GSH or carnosine, 0.25 mM bovine beta-lactoglobulin (beta-LG), and 100 mM phosphate buffer (pH 7.4, 10% ethanol) for 24 h at 37 degrees C, virtually every Lys of beta-LG was found to be fractionally cross-linked to GSH. Cross-linking of Lys to carnosine was less efficient. Using cytochrome c and RNase A, we showed that ONE becomes more protein-reactive in the presence of GSH, whereas protein modification by 4-hydroxy-2-nonenal is inhibited by GSH. Stable antielectrophile-ONE-protein cross-links may serve as biomarkers of oxidative stress and may represent a novel mechanism of irreversible protein glutathionylation.
Collapse
Affiliation(s)
- Xiaochun Zhu
- Departments of Chemistry, Pharmacology, and Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
Biotransformation is essential to convert lipophilic chemicals to water-soluble and readily excretable metabolites. Formally, biotransformation reactions are classified into phase I and phase II reactions. Phase I reactions represent the introduction of functional groups, whereas phase II reactions are conjugations of such functional groups with endogenous, polar products. Biotransformation also plays an essential role in the toxicity of many chemicals due to the metabolic formation of toxic metabolites. These may be classified as stable but toxic products, reactive electrophiles, radicals, and reactive oxygen metabolites. The interaction of toxic products formed by biotransformation reactions with cellular macromolecules initiates the sequences resulting in cellular damage, cell death and toxicity.
Collapse
Affiliation(s)
- Wolfgang Dekant
- Department of Toxicology, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
25
|
Dinkova-Kostova A. The Isothiocyanate Sulforaphane Induces the Phase 2 Response by Signaling of the Keap1–Nrf2–ARE Pathway. OXIDATIVE STRESS AND DISEASE 2008. [DOI: 10.1201/9780849381492.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
26
|
Murphy PJ. The development of drug metabolism research as expressed in the publications of ASPET: Part 3, 1984-2008. Drug Metab Dispos 2008; 36:1977-82. [PMID: 18635745 DOI: 10.1124/dmd.108.023226] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The dramatic changes in drug metabolism research in the last 25 years are well documented in the publications of the American Society for Pharmacology and Experimental Therapeutics (ASPET). New analytical tools combined with modern molecular biological techniques have provided unprecedented access to the workings of the cell. A field that concentrated on only a handful of primary enzymes now has a list of hundreds in its purview. Genetic variation, environmental impact, and molecular diversity have all come under study in attempts to follow the fate of drugs and chemicals. Examples from ASPET journals will be used to illustrate the dramatic advancements in the field.
Collapse
Affiliation(s)
- Patrick J Murphy
- College of Pharmacy and Health Sciences, Butler University, Indianapolis, Indiana, USA.
| |
Collapse
|
27
|
Kurtovic S, Shokeer A, Mannervik B. Diverging catalytic capacities and selectivity profiles with haloalkane substrates of chimeric alpha class glutathione transferases. Protein Eng Des Sel 2008; 21:329-41. [DOI: 10.1093/protein/gzn010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
28
|
Vlaykova T, Miteva L, Gulubova M, Stanilova S. Ile105Val GSTP1 polymorphism and susceptibility to colorectal carcinoma in Bulgarian population. Int J Colorectal Dis 2007; 22:1209-15. [PMID: 17404745 DOI: 10.1007/s00384-007-0305-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2007] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Etiologically, the sporadic colorectal cancer (CRC) is a complex and multifactorial disease that is linked to both exogenic and endogenic factors. Accumulating evidence indicates that susceptibility to cancers, including CRC, is mediated by genetically determined differences in the effectiveness of detoxification of potential carcinogens. A member of the glutathione-S-transferase (GST) family, GSTP1, is an important candidate for involvement in susceptibility to carcinogen-associated CRC. An A-->G transition in exon 5 of the GSTP1 gene resulting in Ile(105)Val amino acid substitution has been identified. This change leads to alteration in catalytic efficiency of variant enzyme. The aim of the current study was to evaluate the influence of Ile(105)Val GSTP1 polymorphism on susceptibility to CRC. MATERIALS AND METHODS The GSTP1 genotyping was conducted in a case-control study of 80 ethnic Bulgarian CRC patients and 126 unaffected controls using polymerase chain reaction restriction fragment length polymorphism method. RESULTS A statistically significant case-control difference in genotype frequencies was observed: 0.69 vs 0.54 for Ile/Ile, 0.22 vs 0.39 for Ile/Val, and 0.09 vs 0.07 for Val/Val (p = 0.049). The odds ratio (OR) for Val/Val was close to 1 (0.96, 95%CI: 0.35-2.66, p = 0.942), whereas the OR for Ile/Val was significantly lower, 0.45 (95%CI: 0.24-0.86, p = 0.016), compared to the referent Ile/Ile genotype. Although a prevalence of the GSTP1 variant allele-containing genotypes (Ile/Val or Val/Val) was found in controls than in patients (OR = 0.53, 95%CI: 0.30-0.96, p = 0.035), the allele frequencies did not show significant difference between cases and controls (p = 0.127). CONCLUSIONS Based on the obtained protective effect of Ile/Val GSTP1 genotype, we could suggest that Ile(105)Val GSTP1 polymorphism may play some role in susceptibility to CRC.
Collapse
Affiliation(s)
- Tatyana Vlaykova
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria.
| | | | | | | |
Collapse
|
29
|
Anders MW. Chemical Toxicology of Reactive Intermediates Formed by the Glutathione-Dependent Bioactivation of Halogen-Containing Compounds. Chem Res Toxicol 2007; 21:145-59. [PMID: 17696489 DOI: 10.1021/tx700202w] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The concept that reactive intermediate formation during the biotransformation of drugs and chemicals is an important bioactivation mechanism was proposed in the 1970s and is now accepted as a major mechanism for xenobiotic-induced toxicity. The enzymology of reactive intermediate formation as well as the characterization of the formation and fate of reactive intermediates are now well-established. The mechanism by which reactive intermediates cause cell damage and death is, however, still poorly understood. Although most xenobiotic-metabolizing enzymes catalyze the bioactivation of chemicals, glutathione-dependent biotransformation has been largely associated with detoxication processes, particularly mercapturic acid formation. Abundant evidence now shows that glutathione-dependent biotransformation constitutes an important bioactivation mechanism for halogen-containing drugs and chemicals and has for many compounds been implicated in their organ-selective toxicity and in their mutagenic and carcinogenic potential. The glutathione-dependent biotransformation of haloalkenes is the first step in the cysteine S-conjugate beta-lyase pathway for the bioactivation of nephrotoxic haloalkenes. This pathway has been a rich source of reactive intermediates, including thioacyl halides, alpha-chloroalkenethiolates, 3-halo-alpha-thiolactones, 2,2,3-trihalothiiranes, halothioketenes, and vinylic sulfoxides. Glutathione-dependent bioactivation of gem-dihalomethanes and 1,2-, 1,3-, and 1,4-dihaloalkanes leads to the formation of alpha-chlorosulfides, thiiranium ions, sulfenate esters, and tetrahydrothiophenium ions, respectively, and these reactions lead to reactive intermediate formation.
Collapse
Affiliation(s)
- M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York 214642, USA
| |
Collapse
|
30
|
Kurtovic S, Jansson R, Mannervik B. Colorimetric endpoint assay for enzyme-catalyzed iodide ion release for high-throughput screening in microtiter plates. Arch Biochem Biophys 2007; 464:284-7. [PMID: 17490601 DOI: 10.1016/j.abb.2007.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 04/04/2007] [Accepted: 04/06/2007] [Indexed: 11/19/2022]
Abstract
Efforts are being made to engineer enzymes with enhanced activities against haloalkanes, a toxicologically important class of compounds widely used and frequently occurring in the environment. Here we describe a facile, inexpensive, and robust method for the screening of libraries of mutated enzymes with iodoalkane substrates. Iodide formed in the enzymatic reaction is oxidized to iodine, which in the presence of starch gives blue color that can be measured at 610nm or scored with the human eye. The assay can be performed with enzymes in crude cell lysates in 96-wells microtiter plates. Expression clones of several glutathione transferases showed diverse activities with different iodoalkanes, and a mutant library of human glutathione transferase A1-1 expressed variants with enhanced substrate selectivities.
Collapse
Affiliation(s)
- Sanela Kurtovic
- Department of Biochemistry and Organic Chemistry, Uppsala University, BMC, Box 576, SE-75123 Uppsala, Sweden
| | | | | |
Collapse
|
31
|
Abstract
Cysteine S-conjugate beta-lyases are pyridoxal 5'-phosphate-containing enzymes that catalyze beta-elimination reactions with cysteine S-conjugates that possess an electron-withdrawing group attached at the sulfur. The end products of the beta-lyase reaction are pyruvate, ammonium and a sulfur-containing fragment. If the sulfur-containing fragment is reactive, the parent cysteine S-conjugate may be toxic, particularly to kidney mitochondria. Halogenated alkenes are examples of electrophiles that are bioactivated (toxified) by conversion to cysteine S-conjugates. These conjugates are converted by cysteine S-conjugate beta-lyases to thioacylating fragments. Several cysteine S-conjugates found in allium foods (garlic and onion) are beta-lyase substrates. This finding may account in part for the chemopreventive activity of allium products. This review (1) identifies enzymes that catalyze cysteine S-conjugate beta-lyase reactions, (2) suggests that toxicant channeling may contribute to halogenated cysteine S-conjugate-induced toxicity to mitochondria, and (3) proposes mechanisms that may contribute to the antiproliferative effects of sulfur-containing fragments eliminated from allium-derived cysteine S-conjugates.
Collapse
Affiliation(s)
- A J L Cooper
- Department of Biochemistry, Weill Medical College, Cornell University, New York, New York, USA.
| | | |
Collapse
|
32
|
Lock EA, Reed CJ. Trichloroethylene: mechanisms of renal toxicity and renal cancer and relevance to risk assessment. Toxicol Sci 2006; 91:313-31. [PMID: 16421178 DOI: 10.1093/toxsci/kfj107] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
1,1,2-Trichloroethylene (TCE) is an important solvent that is widespread in the environment. We have reviewed carcinogenicity data from seven bioassays with regard to renal injury and renal tumors. We report a consistent but low incidence of renal tubule carcinoma in male rats. Epidemiology studies on workers exposed to TCE (and other chlorinated solvents) indicate a weak association between high-level exposure and renal cancer. There appears to be a threshold below which no renal injury or carcinogenicity is expected to arise. TCE is not acutely nephrotoxic to rats or mice, but subchronic exposure to rats produces a small increase in urinary markers of renal injury. Following chronic exposure, pathological changes (toxic nephrosis and a high incidence of cytomegaly and karyomegaly) were observed. The basis for the chronic renal injury probably involves bioactivation of TCE. Based on the classification by E. A. Lock and G. C. Hard (2004, Crit. Rev. Toxicol. 34, 211-299) of chemicals that induce renal tubule tumors, we found no clear evidence to place TCE in category 1 or 2 (chemicals that directly or indirectly interact with renal DNA), category 4 (direct cytotoxicity and sustained tubule cell regeneration), category 5 (indirect cytotoxicity and sustained tubule cell regeneration associated with alpha2u-globulin accumulation), or category 6 (exacerbation of spontaneous chronic progressive nephropathy). TCE is best placed in category 3, chemicals that undergo conjugation with GSH and subsequent enzymatic activation to a reactive species. The implication for human risk assessment is that TCE should not automatically be judged by linear default methods; benchmark methodology could be used.
Collapse
Affiliation(s)
- Edward A Lock
- School of Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF United Kingdom.
| | | |
Collapse
|
33
|
Kharasch ED, Schroeder JL, Bammler T, Beyer R, Srinouanprachanh S. Gene expression profiling of nephrotoxicity from the sevoflurane degradation product fluoromethyl-2,2-difluoro-1-(trifluoromethyl)vinyl ether ("compound A") in rats. Toxicol Sci 2005; 90:419-31. [PMID: 16384817 DOI: 10.1093/toxsci/kfj088] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The major degradation product of the volatile anesthetic sevoflurane, the haloalkene fluoromethyl-2,2-difluoro-1-(trifluoromethyl)vinyl ether (FDVE or "compound A"), is nephrotoxic in rats. FDVE undergoes complex metabolism and bioactivation, which mediates the nephrotoxicity. Nevertheless, the molecular and cellular mechanisms of FDVE toxification are unknown. This investigation evaluated the gene expression profile of kidneys in rats administered a nephrotoxic dose of FDVE. Male Fischer 344 rats (five per group) received 0.25 mmol/kg intraperitoneal FDVE or corn oil (controls) and were sacrificed after 24 or 72 h. Urine output and kidney histological changes were quantified. Kidney RNA was extracted for microarray analysis using Affymetrix GeneChip Rat Expression Array 230A arrays. Quantitative real-time PCR confirmed the modulation of several genes. FDVE caused significant diuresis and necrosis at 24 h, with normal urine output and evidence of tubular regeneration at 72 h. There were 517 informative genes that were differentially expressed >1.5-fold (p < 0.05) versus control at 24 h, of which 283 and 234 were upregulated and downregulated, respectively. Major classes of upregulated genes included those involved in apoptosis, oxidative stress, and inflammatory response (mostly at 24 h), and regeneration and repair; downregulated genes were generally associated with transporters and intermediary metabolism. Among the quantitatively most upregulated genes were kidney injury molecule, osteopontin, clusterin, tissue inhibitor of metalloproteinase 1, and TNF receptor 12, which have been associated with other forms of nephrotoxicity, and angiopoietin-like protein 4, glycoprotein nmb, ubiquitin hydrolase, and HSP70. Microarray results were confirmed by quantitative real-time PCR. FDVE causes rapid and brisk changes in gene expression, providing potential insights into the mechanism of FDVE toxification, and potential biomarkers for FDVE nephrotoxicity which are more sensitive than conventional measures of renal function.
Collapse
Affiliation(s)
- Evan D Kharasch
- Department of Anesthesiology, Washington University, St. Louis, Missouri 63110-1093, USA.
| | | | | | | | | |
Collapse
|
34
|
Bicker W, Lämmerhofer M, Genser D, Kiss H, Lindner W. A case study of acute human chlorpyrifos poisoning: Novel aspects on metabolism and toxicokinetics derived from liquid chromatography–tandem mass spectrometry analysis of urine samples. Toxicol Lett 2005; 159:235-51. [PMID: 15996837 DOI: 10.1016/j.toxlet.2005.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 05/23/2005] [Accepted: 05/25/2005] [Indexed: 01/19/2023]
Abstract
The metabolic fate of the organophosphorothioate-type insecticide chlorpyrifos (CP) in an acutely intoxicated 59 years old female was investigated by liquid chromatography-electrospray ionisation-tandem mass spectrometry (LC-ESI-MS/MS) analysis of urine samples. Fifteen metabolites of CP and its bioactivated intermediate chlorpyrifos-oxon (CPO), respectively, of which only three have been described in man so far, were identified on the basis of characteristic MS/MS transitions, precursor/product ion and/or neutral loss scans, chlorine isotopomer patterns, and partly by synthesis of reference compounds and subsequent structure confirmation. Three distinct biotransformation routes of CP are proposed: (1) cleavage reactions at the aromatic phosphoester bond, (2) cleavage reactions at the alkyl phosphoester bonds, and (3) glutathione (GSH) dependent nucleophilic substitution of the 6-chlorine at the aromatic moiety. Route (2) has not been reported in humans before and (3) is a hitherto completely unknown scheme of CP metabolism. Urinary markers of the latter were chiefly cysteine S-conjugates of mono-dechlorinated CP, CPO, mono-O-deethyl CP, and mono-O-deethyl CPO as well as the 6-mercapturic acid conjugate of 3,5-dichloro-2-pyridinol. The presence of 3,5-dichloro-6-methylthio-2-pyridinol as well as its O-glucuronide suggests further a cysteine S-conjugate beta-lyase mediated degradation. In addition to the qualitative LC-MS/MS screening the renal elimination profiles of the primary products of scheme (1), i.e. diethyl thiophosphate (DETP), diethyl phosphate (DEP), and 3,5,6-trichloro-2-pyridinol (TCP), were monitored over 14 days (n=21). A biphasic first-order excretion mechanism with half-lives of 21.5h (initial fast excretion phase) and 119.5h (terminal phase) for the sum of free DETP and DEP was found. TCP was hardly eliminated in its free form (O-glucuronide identified as phase II conjugate) and half-lives calculated for the total amount of TCP (acidic hydrolysis of urine samples) were 40.8 and 150.7h. The present study gives a more detailed view on the biotransformation of CP and together with the obtained kinetic data adds novel aspects to the limited knowledge of human metabolism of this xenobiotic, in particular at high dosage.
Collapse
Affiliation(s)
- Wolfgang Bicker
- Christian Doppler Laboratory for Molecular Recognition Materials, Department of Analytical and Food Chemistry, University of Vienna, Währinger Strasse 38, A-1090 Vienna, Austria
| | | | | | | | | |
Collapse
|
35
|
Abstract
Thioredoxin (Trx) is a redox-active protein that has been shown to regulate various cellular processes due to its thiol-disulfide exchange reaction. It has antioxidant properties and also induces the expression of critical antioxidant enzymes such as manganese superoxide dismutase. Trx along with thioredoxin reductase and peroxiredoxins forms a complete system similar to the glutathione system, but with different and divergent functions. This review is a mini-update on key advances in the role of Trx in signal transduction and premature newborn biology. In addition, this mini-update also reviews recently reported prooxidant properties of Trx that relate to anthracycline redox cycling.
Collapse
Affiliation(s)
- Kumuda C Das
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR7205, USA.
| |
Collapse
|