1
|
Potočnjak I, Marinić J, Batičić L, Šimić L, Broznić D, Domitrović R. Aucubin administered by either oral or parenteral route protects against cisplatin-induced acute kidney injury in mice. Food Chem Toxicol 2020; 142:111472. [DOI: 10.1016/j.fct.2020.111472] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023]
|
2
|
Nephroprotective Effect of Herbal Extract Eurycoma longifolia on Paracetamol-Induced Nephrotoxicity in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4916519. [PMID: 31214269 PMCID: PMC6535855 DOI: 10.1155/2019/4916519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/26/2019] [Accepted: 03/31/2019] [Indexed: 11/18/2022]
Abstract
Paracetamol (PCM) is a well-known drug widely used for its analgesic and antipyretic properties. PCM is generally considered as safe but overdose of PCM can cause nephrotoxicity. Traditionally, herbs have been used for the treatment of drug or toxin-induced renal disorders and numerous medicinal plants were tested for nephroprotection effect in PCM-induced nephrotoxicity model. The aim of the present study was to evaluate the protective effect of the herbal extract Eurycoma longifolia (EL) against PCM-induced nephrotoxicity rat model. Forty Wistar rats were randomly divided into five groups of eight rats each: control (vehicle 10 ml/kg), PCM alone (200 mg/kg PCM), EL 100 (EL 100 mg/kg+200 mg/kg PCM), EL 200 (EL 200 mg/kg+200 mg/kg PCM), and EL 400 (EL 400 mg/kg+200 mg/kg PCM). All animals from control group received vehicle daily and animals from groups PCM alone, EL 100, EL 200, and EL 400 received repeated dose of PCM and the assigned treatment of EL daily for a period of 14 days. On the 15th day, serum creatinine, blood urea nitrogen, protein, and albumin were measured in blood and creatinine clearance was measured in urine collected over 24 hours. Kidney sections of all experimental groups underwent histopathological analysis. There was a significant (p<0.05) increase in serum creatinine and blood urea levels in the PCM alone group compared to the treatment groups due to nephrotoxicity. In the treatment groups, there was a dose-dependent protection against PCM-induced changes observed in serum total protein, albumin, urea, and creatinine. Significant (p<0.05) drop was seen in serum creatinine and blood urea content in EL 200 and EL 400 groups. Creatinine clearance significantly increased for EL 200 (p<0.01) and EL 400 (p < 0.001) groups. Serum total protein and serum albumin content were significantly increased (p<0.05) in EL 200 and EL 400 groups compared to PCM alone group. Histopathological examination (H&E staining) of the rat kidneys revealed severe degeneration in the PCM alone group, while there was evidence of significant dose-dependent protection in the treatment groups against PCM-induced changes. The serum and urine biochemical results and histopathology analysis of the kidney indicate the nephroprotective potential of EL extract against PCM-induced nephrotoxicity.
Collapse
|
3
|
Wang X, Xin X, Sun Y, Zou L, Li H, Zhao Y, Li R, Peng Y, Zheng J. Chemical Reactivity of Aloe-Emodin and Its Hydroxylation Metabolites to Thiols. Chem Res Toxicol 2019; 32:234-244. [DOI: 10.1021/acs.chemrestox.8b00248] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xu Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Xin Xin
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Ying Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Lizhu Zou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Hui Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yufei Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Ruihong Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
4
|
Lin D, Kostov R, Huang JTJ, Henderson CJ, Wolf CR. Novel Pathways of Ponatinib Disposition Catalyzed By CYP1A1 Involving Generation of Potentially Toxic Metabolites. J Pharmacol Exp Ther 2017; 363:12-19. [PMID: 28882992 PMCID: PMC5596814 DOI: 10.1124/jpet.117.243246] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023] Open
Abstract
Ponatinib, a pan-BCR-ABL tyrosine kinase inhibitor for the treatment of chronic myeloid leukemia (CML), causes severe side effects including vascular occlusions, pancreatitis, and liver toxicity, although the underlying mechanisms remain unclear. Modifications of critical proteins through reactive metabolites are thought to be responsible for a number of adverse drug reactions. In vitro metabolite screening of ponatinib with human liver microsomes and glutathione revealed unambiguous signals of ponatinib-glutathione (P-GSH) adducts. Further profiling of human cytochrome P450 (P450) indicated that CYP1A1 was the predominant P450 enzyme driving this reaction. P-GSH conjugate formation paralleled the disappearance of hydroxylated ponatinib metabolites, suggesting the initial reaction was epoxide generation. Mouse glutathione S-transferase p1 (mGstp1) further enhanced P-GSH adduct formation in vitro. Ponatinib pharmacokinetics were determined in vivo in wild-type (WT) mice and mice humanized for CYP1A1/2 and treated with the CYP1A1 inducers 2,3,7,8-tetrachlorodibenzodioxin or 3-methylcholanthrene. Ponatinib exposure was significantly decreased in treated mice compared with controls (7.7- and 2.2-fold for WT and humanized CYP1A1/2, respectively). Interestingly, the P-GSH conjugate was only found in the feces of CYP1A1-induced mice, but not in control animals. Protein adducts were also identified by liquid chromatography-tandem mass spectrometry analysis of mGstp1 tryptic digests. These results indicate that not only could CYP1A1 be involved in ponatinib disposition, which has not been previously reported, but also that electrophilic intermediates resulting from CYP1A1 metabolism in normal tissues may contribute to ponatinib toxicity. These data are consistent with a recent report that CML patients who smoke are at greater risk of disease progression and premature death.
Collapse
Affiliation(s)
- De Lin
- Division of Cancer Research, Jacqui Wood Cancer Centre (D.L., C.J.H., C.R.W.), Molecular & Cellular Medicine (R.K.), and Biomarker & Drug Analysis Core (J.T.-J.H.), School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Rumen Kostov
- Division of Cancer Research, Jacqui Wood Cancer Centre (D.L., C.J.H., C.R.W.), Molecular & Cellular Medicine (R.K.), and Biomarker & Drug Analysis Core (J.T.-J.H.), School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Jeffrey T-J Huang
- Division of Cancer Research, Jacqui Wood Cancer Centre (D.L., C.J.H., C.R.W.), Molecular & Cellular Medicine (R.K.), and Biomarker & Drug Analysis Core (J.T.-J.H.), School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Colin J Henderson
- Division of Cancer Research, Jacqui Wood Cancer Centre (D.L., C.J.H., C.R.W.), Molecular & Cellular Medicine (R.K.), and Biomarker & Drug Analysis Core (J.T.-J.H.), School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - C Roland Wolf
- Division of Cancer Research, Jacqui Wood Cancer Centre (D.L., C.J.H., C.R.W.), Molecular & Cellular Medicine (R.K.), and Biomarker & Drug Analysis Core (J.T.-J.H.), School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
5
|
Horii I. The principle of safety evaluation in medicinal drug - how can toxicology contribute to drug discovery and development as a multidisciplinary science? J Toxicol Sci 2017; 41:SP49-SP67. [PMID: 28250284 DOI: 10.2131/jts.41.sp49] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Pharmaceutical (drug) safety assessment covers a diverse science-field in the drug discovery and development including the post-approval and post-marketing phases in order to evaluate safety and risk management. The principle in toxicological science is to be placed on both of pure and applied sciences that are derived from past/present scientific knowledge and coming new science and technology. In general, adverse drug reactions are presented as "biological responses to foreign substances." This is the basic concept of thinking about the manifestation of adverse drug reactions. Whether or not toxic expressions are extensions of the pharmacological effect, adverse drug reactions as seen from molecular targets are captured in the category of "on-target" or "off-target", and are normally expressed as a biological defense reaction. Accordingly, reactions induced by pharmaceuticals can be broadly said to be defensive reactions. Recent molecular biological conception is in line with the new, remarkable scientific and technological developments in the medical and pharmaceutical areas, and the viewpoints in the field of toxicology have shown that they are approaching toward the same direction as well. This paper refers to the basic concept of pharmaceutical toxicology, the differences for safety assessment in each stage of drug discovery and development, regulatory submission, and the concept of scientific considerations for risk assessment and management from the viewpoint of "how can multidisciplinary toxicology contribute to innovative drug discovery and development?" And also realistic translational research from preclinical to clinical application is required to have a significant risk management in post market by utilizing whole scientific data derived from basic and applied scientific research works. In addition, the significance for employing the systems toxicology based on AOP (Adverse Outcome Pathway) analysis is introduced, and coming challenges on precision medicine are to be addressed for the new aspect of efficacy and safety evaluation.
Collapse
Affiliation(s)
- Ikuo Horii
- Global Drug Safety Research & Development, Pfizer
| |
Collapse
|
6
|
Feng Y, Wang H, Wang Q, Huang W, Peng Y, Zheng J. Chemical Interaction of Protein Cysteine Residues with Reactive Metabolites of Methyleugenol. Chem Res Toxicol 2017; 30:564-573. [DOI: 10.1021/acs.chemrestox.6b00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yukun Feng
- Wuya
College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Hui Wang
- Wuya
College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Qian Wang
- Wuya
College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Wenlin Huang
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Ying Peng
- Wuya
College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jiang Zheng
- Wuya
College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| |
Collapse
|
7
|
Zhang H, Gan J, Shu YZ, Humphreys WG. High-Resolution Mass Spectrometry-Based Background Subtraction for Identifying Protein Modifications in a Complex Biological System: Detection of Acetaminophen-Bound Microsomal Proteins Including Argininosuccinate Synthetase. Chem Res Toxicol 2015; 28:775-81. [DOI: 10.1021/tx500526s] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Haiying Zhang
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - Jinping Gan
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - Yue-Zhong Shu
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - W. Griffith Humphreys
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| |
Collapse
|
8
|
Yang Y, Xiao Q, Humphreys WG, Dongre A, Shu YZ. Identification of Human Liver Microsomal Proteins Adducted by a Reactive Metabolite Using Shotgun Proteomics. Chem Res Toxicol 2014; 27:1537-46. [DOI: 10.1021/tx500181p] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Yanou Yang
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Qing Xiao
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - W. Griffith Humphreys
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Ashok Dongre
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Yue-Zhong Shu
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| |
Collapse
|
9
|
Schneider KJ, DeCaprio AP. Covalent Thiol Adducts Arising from Reactive Intermediates of Cocaine Biotransformation. Chem Res Toxicol 2013; 26:1755-64. [DOI: 10.1021/tx4003116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kevin J. Schneider
- Department of Chemistry and
Biochemistry and the International Forensic Research Institute, Florida International University, 11200 SW Eighth Street, Miami, Florida 33199, United States
| | - Anthony P. DeCaprio
- Department of Chemistry and
Biochemistry and the International Forensic Research Institute, Florida International University, 11200 SW Eighth Street, Miami, Florida 33199, United States
| |
Collapse
|
10
|
Patlewicz G, Ball N, Booth ED, Hulzebos E, Zvinavashe E, Hennes C. Use of category approaches, read-across and (Q)SAR: General considerations. Regul Toxicol Pharmacol 2013; 67:1-12. [DOI: 10.1016/j.yrtph.2013.06.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/25/2013] [Accepted: 06/03/2013] [Indexed: 10/26/2022]
|
11
|
Yuan W, Jin H, Chung JK, Zheng J. Evidence for cellular protein covalent binding derived from styrene metabolite. Chem Biol Interact 2010; 186:323-30. [PMID: 20470765 PMCID: PMC3463232 DOI: 10.1016/j.cbi.2010.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 05/06/2010] [Accepted: 05/06/2010] [Indexed: 12/13/2022]
Abstract
Styrene is one of the most important industrial intermediates consumed in the world. Human exposure to styrene occurs mainly in the reinforced plastics industry, particularly in developing countries. Styrene has been found to be hepatotoxic and pneumotoxic in humans and animals. The biochemical mechanisms of styrene-induced toxicities remain unknown. Albumin and hemoglobin adduction derived from styrene oxide, a major reactive metabolite of styrene, has been reported in blood samples obtained from styrene-exposed workers. The objectives of the current study focused on cellular protein covalent binding of styrene metabolite and its correlation with cytotoxicity induced by styrene. We found that radioactivity was bound to cellular proteins obtained from mouse airway trees after incubation with (14)C-styrene. Microsomal incubation studies showed that the observed protein covalent binding required the metabolic activation of styrene. The observed radioactivity binding in protein samples obtained from the cultured airways and microsomal incubations was significantly suppressed by co-incubation with disulfiram, a CYP2E1 inhibitor, although disulfiram apparently did not show a protective effect against the cytotoxicity of styrene. A 2-fold increase in radioactivity bound to cellular proteins was detected in cells stably transfected with CYP2E1 compared to the wild-type cells after (14)C-styrene exposure. With the polyclonal antibody developed in our lab, we detected cellular protein adduction derived from styrene oxide at cysteinyl residues in cells treated with styrene. Competitive immunoblot studies confirmed the modification of cysteine residues by styrene oxide. Cell culture studies showed that the styrene-induced protein modification and cell death increased with the increasing concentration of styrene exposure. In conclusion, we detected cellular protein covalent modification by styrene oxide in microsomal incubations, cultured cells, and mouse airways after exposure to styrene and found a good correlation between styrene-induced cytotoxicity and styrene oxide-derived cellular protein adduction.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Medicine, University of Washington, Seattle, WA 98195
| | - Hua Jin
- Center for Developmental Therapeutics, Seattle Children’s Research Institute, Division of Gastroenterology, Department of Pediatrics, University of Washington, Seattle, WA 98101
| | - Jou-Ku Chung
- Department of Drug Metabolism and Pharmacokinetics, Archemix Corp., Cambridge, MA 02142
| | - Jiang Zheng
- Center for Developmental Therapeutics, Seattle Children’s Research Institute, Division of Gastroenterology, Department of Pediatrics, University of Washington, Seattle, WA 98101
| |
Collapse
|
12
|
Saquib Q, Al-Khedhairy AA, Alarifi SA, Dwivedi S, Mustafa J, Musarrat J. Fungicide methyl thiophanate binding at sub-domain IIA of human serum albumin triggers conformational change and protein damage. Int J Biol Macromol 2010; 47:60-7. [DOI: 10.1016/j.ijbiomac.2010.03.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 03/06/2010] [Accepted: 03/26/2010] [Indexed: 10/19/2022]
|
13
|
Hampel M, Alonso E, Aparicio I, Bron JE, Santos JL, Taggart JB, Leaver MJ. Potential physiological effects of pharmaceutical compounds in Atlantic salmon (Salmo salar) implied by transcriptomic analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2010; 17:917-933. [PMID: 20140651 DOI: 10.1007/s11356-009-0282-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 12/13/2009] [Indexed: 05/28/2023]
Abstract
BACKGROUND, AIM, AND SCOPE Pharmaceuticals are emerging pollutants widely used in everyday urban activities which can be detected in surface, ground, and drinking waters. Their presence is derived from consumption of medicines, disposal of expired medications, release of treated and untreated urban effluents, and from the pharmaceutical industry. Their growing use has become an alarming environmental problem which potentially will become dangerous in the future. However, there is still a lack of knowledge about long-term effects in non-target organisms as well as for human health. Toxicity testing has indicated a relatively low acute toxicity to fish species, but no information is available on possible sublethal effects. This study provides data on the physiological pathways involved in the exposure of Atlantic salmon as representative test species to three pharmaceutical compounds found in ground, surface, and drinking waters based on the evaluation of the xenobiotic-induced impairment resulting in the activation and silencing of specific genes. MATERIALS AND METHODS Individuals of Atlantic salmon (Salmo salar) parr were exposed during 5 days to environmentally relevant concentrations of three representative pharmaceutical compounds with high consumption rates: the analgesic acetaminophen (54.77+/-34.67 microg L(-1)), the anticonvulsant carbamazepine (7.85+/-0.13 microg L(-1)), and the beta-blocker atenolol (11.08+/-7.98 microg L(-1)). Five immature males were selected for transcriptome analysis in brain tissues by means of a 17k salmon cDNA microarray. For this purpose, mRNA was isolated and reverse-transcribed into cDNA which was labeled with fluorescent dyes and hybridized against a common pool to the arrays. Lists of significantly up- and down-regulated candidate genes were submitted to KEGG (Kyoto Encyclopedia of Genes and Genomes) in order to analyze for induced pathways and to evaluate the usefulness of this method in cases of not completely annotated test organisms. RESULTS Exposure during 5 days to environmentally relevant concentrations of the selected pharmaceutical compounds acetaminophen, carbamazepine, and atenolol produced differences in the expression of 659, 700, and 480 candidate genes, respectively. KEGG annotation numbers (KO annotations) were obtained for between 26.57% and 33.33% of these differently expressed genes per treatment in comparison to non-exposure conditions. Pathways that showed to be induced did not always follow previously reported targets or metabolic routes for the employed treatments; however, several other pathways have been found (four or more features) to be significantly induced. DISCUSSION Energy-related pathways have been altered under exposure in all the selected treatments, indicating a possible energy budget leakage due to additional processes resulting from the exposure to environmental contaminants. Observed induction of pathways may indicate additional processes involved in the mode of action of the selected pharmaceuticals which may not have been detected with conventional methods like quantitative PCR in which only suspected features are analyzed punctually for effects. The employment of novel high-throughput screening techniques in combination with global pathway analysis methods, even if the organism is not completely annotated, allows the examination of a much broader range of candidates for potential effects of exposure at the gene level. CONCLUSIONS The continuously growing number of annotations of representative species relevant for environmental quality testing is facilitating pathway analysis processes for not completely annotated organisms. KEGG has shown to be a useful tool for the analysis of induced pathways from data generated by microarray techniques with the selected pharmaceutical contaminants acetaminophen, carbamazepine, and atenolol, but further studies have to be carried out in order to determine if a similar expression pattern in terms of fold change quantity and pathways is observed after long-term exposure. Together with the information obtained in this study, it will then be possible to evaluate the potential risk that the continuous release of these compounds may have on the environment and ecosystem functioning.
Collapse
Affiliation(s)
- Miriam Hampel
- Institute of Aquaculture, University of Stirling, FK9 4LA, Stirling, UK.
| | | | | | | | | | | | | |
Collapse
|
14
|
Giri S, Nieber K, Bader A. Hepatotoxicity and hepatic metabolism of available drugs: current problems and possible solutions in preclinical stages. Expert Opin Drug Metab Toxicol 2010; 6:895-917. [DOI: 10.1517/17425251003792521] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
15
|
Palani S, Raja S, Naresh R, Senthil Kumar B. Evaluation of nephroprotective, diuretic, and antioxidant activities ofplectranthus amboinicuson acetaminophen-induced nephrotoxic rats. Toxicol Mech Methods 2010; 20:213-21. [DOI: 10.3109/15376511003736787] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
16
|
Horii I. Toxic effect onset and evaluations of medicinal drugs - horizon for Darwinian toxicological thought -. J Toxicol Sci 2010; 35:425-35. [DOI: 10.2131/jts.35.425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Ikuo Horii
- Showa University (Department of Biochemical Toxicology, School of Pharmaceutical Sciences)
- Pfizer (Global Research & Development, Drug Safety Research & Development)
| |
Collapse
|
17
|
Abstract
From the studies that have been done by many laboratories over the last 2 decades, it is now clear that the toxicities produced by many drugs are due to their reactive metabolites. It is though that, in many cases, reactive metabolites cause toxicity by binding covalently to tissue proteins. However, until recently it was difficult to identify these protein targets. Due to the development of an immunochemical approach, this problem has been overcome, as is illustrated here by studies that have been conducted on the metabolic basis of the idiosyncratic hepatitis caused by the inhalation anaesthetic halothane. The major problem to solve in the future will be to determine how protein adduct formation leads to toxicity. It is possible that protein adduct formation may alter an important cellular function or may lead to immunopathology, as is thought to occur in the case of halothane hepatitis. If an allergic reaction is suspected, purified protein targets of reactive metabolites can serve as antigens for identifying sensitized individuals. This information can be used to prevent not only an allergic reaction to the drug, but possible cross-reactions to other drugs that are structurally related. Another important application of these studies is the design of safer alternative drugs that will not produce structurally similar toxic reactive metabolites.
Collapse
Affiliation(s)
- L R Pohl
- Molecular and Cellular Toxicology Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1760, USA.
| | | | | |
Collapse
|
18
|
Lohmann W, Hayen H, Karst U. Covalent Protein Modification by Reactive Drug Metabolites Using Online Electrochemistry/Liquid Chromatography/Mass Spectrometry. Anal Chem 2008; 80:9714-9. [DOI: 10.1021/ac801699g] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Wiebke Lohmann
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| | - Heiko Hayen
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| | - Uwe Karst
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW To examine recent advances in our understanding of how drugs can trigger a hypersensitivity reaction in the liver, how tolerance is lost, the mechanisms of damage to hepatocytes and the strategies towards a better assessment of an idiosyncratic drug liver reaction. RECENT FINDINGS Formation and presentation of drug-protein adducts, or a direct interaction with the major histocompatibility complex/T-cell receptor complex is a necessary but not sufficient stimulus to trigger a hypersensitivity reaction. Liver shows considerable tolerogenic potential towards drug adducts. Recent studies highlight allergic hepatitis as a loss of liver tolerance towards drug antigens, the mechanisms of which are beginning to be unravelled. Cell injury caused by the drug itself, a concomitant inflammatory process, or a coincidental stimulus probably represents the additional signal needed to initiate the allergic process. SUMMARY Drug-induced liver injury is of concern due to its unpredictable nature and serious clinical implications. Clinically, both hepatocellular injury and cholestasis can occur and most episodes have good clinical prognoses upon drug discontinuation. In a few cases, damage to the liver cells may continue in the form of an autoimmune hepatitis. The available diagnostic tools to confirm an immune-mediated hepatic injury are still very limited, and rely on the lymphocyte transformation test.
Collapse
Affiliation(s)
- José V Castell
- Unit for Experimental Hepatology, Research Centre, University Hospital La Fe, Spain.
| | | |
Collapse
|
20
|
Welch KD, Reilly TP, Bourdi M, Hays T, Pise-Masison CA, Radonovich MF, Brady JN, Dix DJ, Pohl LR. Genomic identification of potential risk factors during acetaminophen-induced liver disease in susceptible and resistant strains of mice. Chem Res Toxicol 2006; 19:223-33. [PMID: 16485898 DOI: 10.1021/tx050285z] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Drug-induced liver disease (DILD) continues to cause significant morbidity and mortality and impair new drug development. Mounting evidence suggests that DILD is a complex, multifactorial disease in which no one factor is likely to be an absolute indicator of susceptibility. As an approach to better understand the multifactorial basis of DILD, we recently compared the hepatic proteomes of mice that were resistant (SJL) and susceptible (C57Bl/6) to APAP-induced liver disease (AILD) wherein we identified potential risk factors and mechanistic pathways responsible for DILD. In this study, we have uncovered additional potential risk factors by comparing hepatic mRNA expression profiles of the same two strains of mice with that of SJLxB6-F1 hybrid (F1) mice, which were found to be of intermediate susceptibility to AILD. Global hepatic gene expression profiling over a 24 h period following APAP treatment revealed elevated patterns in the mRNA expression of cytoprotective genes in resistant SJL mice as compared to susceptible B6 mice, while F1 mice had intermediate mRNA expression levels of these genes. One of these genes encoded for heat shock protein (HSP) 70 whose relative protein expression among the three strains of mice was found to parallel that of their mRNA levels, suggesting that this protein had a protective role against AILD. However, there was no difference in the susceptibility of HSP70 knockout (KO) mice to AILD as compared to wild-type (WT) mice. There were also protoxicant genes, such as osteopontin (OPN), with elevated mRNA expression levels in the B6 mice as compared to the SJL mice and with intermediate levels in the F1 mice, suggesting that they may play a role in exacerbating liver injury after APAP treatment. In support of this hypothesis, OPN KO mice were found to be more resistant to AILD than WT mice. Additionally, the results from both the proteomic and the genomic studies were compared. The two approaches were found to be complementary to each other and not simply overlapping. Our findings suggest that comparative gene expression analysis of susceptible and resistant mouse strains may lead to the identification of factors that could have a role in determining the susceptibility of individuals to DILD.
Collapse
Affiliation(s)
- Kevin D Welch
- Molecular and Cellular Toxicology Section, Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
GÓMEZ-LECHÓN MJ, CARRASQUER J, BERENGUER J, CASTELL JV. Evidence of antibodies to erythromycin in serum of a patient following an episode of acute drug-induced hepatitis. Clin Exp Allergy 2006. [DOI: 10.1111/j.1365-2222.1996.tb00581.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
22
|
Yang XX, Hu ZP, Chan SY, Zhou SF. Monitoring drug-protein interaction. Clin Chim Acta 2005; 365:9-29. [PMID: 16199025 DOI: 10.1016/j.cca.2005.08.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 08/16/2005] [Accepted: 08/23/2005] [Indexed: 11/25/2022]
Abstract
A variety of therapeutic drugs can undergo biotransformation via Phase I and Phase II enzymes to reactive metabolites that have intrinsic chemical reactivity toward proteins and cause potential organ toxicity. A drug-protein adduct is a protein complex that forms when electrophilic drugs or their reactive metabolite(s) covalently bind to a protein molecule. Formation of such drug-protein adducts eliciting cellular damages and immune responses has been a major hypothesis for the mechanism of toxicity caused by numerous drugs. The monitoring of protein-drug adducts is important in the kinetic and mechanistic studies of drug-protein adducts and establishment of dose-toxicity relationships. The determination of drug-protein adducts can also provide supportive evidence for diagnosis of drug-induced diseases associated with protein-drug adduct formation in patients. The plasma is the most commonly used matrix for monitoring drug-protein adducts due to its convenience and safety. Measurement of circulating antibodies against drug-protein adducts may be used as a useful surrogate marker in the monitoring of drug-protein adducts. The determination of plasma protein adducts and/or relevant antibodies following administration of several drugs including acetaminophen, dapsone, diclofenac and halothane has been conducted in clinical settings for characterizing drug toxicity associated with drug-protein adduct formation. The monitoring of drug-protein adducts often involves multi-step laboratory procedure including sample collection and preliminary preparation, separation to isolate or extract the target compound from a mixture, identification and determination. However, the monitoring of drug-protein adducts is often difficult because of short half-lives of the protein adducts, sampling problem and lack of sensitive analytical techniques for the protein adducts. Currently, chromatographic (e.g. high performance liquid chromatography) and immunological methods (e.g. enzyme-linked immunosorbent assay) are two major techniques used to determine protein adducts of drugs in patients. The present review highlights the importance for clinical monitoring of drug-protein adducts, with an emphasis on methodology and with a further discussion of the application of these techniques to individual drugs and their target proteins.
Collapse
Affiliation(s)
- Xiao-Xia Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Science Drive 4, Singapore 117543, Singapore
| | | | | | | |
Collapse
|
23
|
Castell JV, Donato MT, Gómez-Lechón MJ. Metabolism and bioactivation of toxicants in the lung. The in vitro cellular approach. ACTA ACUST UNITED AC 2005; 57 Suppl 1:189-204. [PMID: 16092727 DOI: 10.1016/j.etp.2005.05.008] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Lung is a target organ for the toxicity of inhalated compounds. The respiratory tract is frequently exposed to elevated concentrations of these compounds and become the primary target site for toxicity. Occupational, accidental or prolonged exposure to a great variety of chemicals may result in acute or delayed injury to cells of the respiratory tract. Nevertheless, lung has a significant capability of biotransforming such compounds with the aim of reducing its potential toxicity. In some instances, the biotransformation of a given compound can result in the generation of more reactive, and frequently more toxic, metabolites. Indeed, lung tissue is known to activate pro-carcinogens (i.e. polycyclic aromatic hydrocarbons or N-nitrosamines) into more reactive intermediates that easily form DNA adducts. Lungs express several enzymes involved in the metabolising of xenobiotics. Among them, cytochrome P450 enzymes are major players in the oxidative metabolism as well metabolic bioactivation of many organic toxicants, including pro-carcinogens. Xenobiotic-metabolising P450 enzymes are expressed in bronchial and bronchiolar epithelium, Clara cells, type II pneumocytes, and alveolar macrophages Individual CYP isoforms have different patterns of localisation within pulmonary tissue. With the aid of sensitive techniques (i.e. reverse transcriptase-polymerase chain reaction, RT-PCR) it has become possible to detect CYP1A1, CYP1B1, CYP2A6, CYP2B6, CYP2E1 and CYP3A5 mRNAs in lung cells. Less conclusive results have been obtained concerning CYP2Cs, CYP2D6 and CYP3A4. CYP3A5 protein appears to be widely present in all lung samples and is localised in the ciliated and mucous cells of the bronchial wall, bronchial glands, bronchiolar ciliated epithelium and in type I and type II alveolar epithelium. Lung cells also express Phase II enzymes such as epoxide hydrolase, UGT1A (glucuronyl transferase) and GST-P1 (glutathione S-transferase), which largely act as detoxifying enzymes. A key question concerning organ-specific chemical toxicity is whether the actual target has the capacity to activate (or efficiently inactivate) chemicals. Results of several studies indicate that the different xenobiotic-metabolising CYPs, present in the human lung and lung-derived cell lines, likely contribute to in situ activation of pulmonary toxins, among them, pro-carcinogens. Some CYPs, in particular CYP1A, are polymorphic and inducible. Interindividual differences in the expression of these CYPs may explain the different risk of developing lung toxicity (possibly cancer), by agents that require metabolic activation. Few cell lines, principally A549, have been used with variable success as an experimental model for investigating the mechanisms of toxicity. Although RT-PCR analysis has evidenced the presence of the major human pulmonary CYP mRNAs, the measurable P450 specific activities are, however, far below those present in human lungs. Detection of the toxicity elicited by reactive metabolites requires the use of metabolically competent cells; consequently, better performing cells are needed to ensure realistic in vitro prediction of toxicity. Genetic manipulation of lung-derived cells allowing them to re-express key biotransformation enzymes appear to be a promising strategy to improve their functionality and metabolic performance.
Collapse
Affiliation(s)
- José V Castell
- Research Center, University Hospital La Fe, Avda de Campanar 21, E-46009 Valencia, Spain.
| | | | | |
Collapse
|
24
|
Zhou S, Chan E, Duan W, Huang M, Chen YZ. Drug bioactivation, covalent binding to target proteins and toxicity relevance. Drug Metab Rev 2005; 37:41-213. [PMID: 15747500 DOI: 10.1081/dmr-200028812] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A number of therapeutic drugs with different structures and mechanisms of action have been reported to undergo metabolic activation by Phase I or Phase II drug-metabolizing enzymes. The bioactivation gives rise to reactive metabolites/intermediates, which readily confer covalent binding to various target proteins by nucleophilic substitution and/or Schiff's base mechanism. These drugs include analgesics (e.g., acetaminophen), antibacterial agents (e.g., sulfonamides and macrolide antibiotics), anticancer drugs (e.g., irinotecan), antiepileptic drugs (e.g., carbamazepine), anti-HIV agents (e.g., ritonavir), antipsychotics (e.g., clozapine), cardiovascular drugs (e.g., procainamide and hydralazine), immunosupressants (e.g., cyclosporine A), inhalational anesthetics (e.g., halothane), nonsteroidal anti-inflammatory drugs (NSAIDSs) (e.g., diclofenac), and steroids and their receptor modulators (e.g., estrogens and tamoxifen). Some herbal and dietary constituents are also bioactivated to reactive metabolites capable of binding covalently and inactivating cytochrome P450s (CYPs). A number of important target proteins of drugs have been identified by mass spectrometric techniques and proteomic approaches. The covalent binding and formation of drug-protein adducts are generally considered to be related to drug toxicity, and selective protein covalent binding by drug metabolites may lead to selective organ toxicity. However, the mechanisms involved in the protein adduct-induced toxicity are largely undefined, although it has been suggested that drug-protein adducts may cause toxicity either through impairing physiological functions of the modified proteins or through immune-mediated mechanisms. In addition, mechanism-based inhibition of CYPs may result in toxic drug-drug interactions. The clinical consequences of drug bioactivation and covalent binding to proteins are unpredictable, depending on many factors that are associated with the administered drugs and patients. Further studies using proteomic and genomic approaches with high throughput capacity are needed to identify the protein targets of reactive drug metabolites, and to elucidate the structure-activity relationships of drug's covalent binding to proteins and their clinical outcomes.
Collapse
Affiliation(s)
- Shufeng Zhou
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
25
|
Gan J, Harper TW, Hsueh MM, Qu Q, Humphreys WG. Dansyl Glutathione as a Trapping Agent for the Quantitative Estimation and Identification of Reactive Metabolites. Chem Res Toxicol 2005; 18:896-903. [PMID: 15892584 DOI: 10.1021/tx0496791] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A sensitive and quantitative method was developed for the estimation of reactive metabolite formation in vitro. The method utilizes reduced glutathione (GSH) labeled with a fluorescence tag as a trapping agent and fluorescent detection for quantitation. The derivatization of GSH was accomplished by reaction of oxidized glutathione (GSSG) with dansyl chloride to form dansylated GSSG. Subsequent reduction of the disulfide bond yielded dansylated GSH (dGSH). Test compounds were incubated with human liver microsomes in the presence of dGSH and NADPH, and the resulting mixtures were analyzed by HPLC coupled with a fluorescence detector and a mass spectrometer for the quantitation and mass determination of the resulting dGSH adducts. The comparative chemical reactivity of dGSH vs GSH was investigated by monitoring the reaction of each with 1-chloro-2,4-dinitrobenzene or R-(+)-pulegone after bioactivation. dGSH was found to be equivalent to GSH in chemical reactivity toward both thiol reactive molecules. dGSH did not serve as a cofactor for glutathione S-transferase (GST)-mediated conjugation of 3,4-dichloronitrobenzene in incubations with either human liver S9 fractions or a recombinant GST, GSTM1-1. Reference compounds were tested in this assay, including seven compounds that have been reported to form GSH adducts along with seven drugs that are among the most prescribed in the current U.S. market and have not been reported to form GSH adducts. dGSH adducts were detected and quantitated in incubations with all seven positive reference compounds; however, there were no dGSH adducts observed with any of the widely prescribed drugs. In comparison with existing methods, this method is sensitive, quantitative, cost effective, and easy to implement.
Collapse
Affiliation(s)
- Jinping Gan
- Department of Pharmaceutical Candidate Optimization, Pharmaceutical Research Institute, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, USA.
| | | | | | | | | |
Collapse
|
26
|
Zhou S. Separation and detection methods for covalent drug–protein adducts. J Chromatogr B Analyt Technol Biomed Life Sci 2003; 797:63-90. [PMID: 14630144 DOI: 10.1016/s1570-0232(03)00399-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Covalent binding of reactive metabolites of drugs to proteins has been a predominant hypothesis for the mechanism of toxicity caused by numerous drugs. The development of efficient and sensitive analytical methods for the separation, identification, quantification of drug-protein adducts have important clinical and toxicological implications. In the last few decades, continuous progress in analytical methodology has been achieved with substantial increase in the number of new, more specific and more sensitive methods for drug-protein adducts. The methods used for drug-protein adduct studies include those for separation and for subsequent detection and identification. Various chromatographic (e.g., affinity chromatography, ion-exchange chromatography, and high-performance liquid chromatography) and electrophoretic techniques [e.g., sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), two-dimensional SDS-PAGE, and capillary electrophoresis], used alone or in combination, offer an opportunity to purify proteins adducted by reactive drug metabolites. Conventionally, mass spectrometric (MS), nuclear magnetic resonance, and immunological and radioisotope methods are used to detect and identify protein targets for reactive drug metabolites. However, these methods are labor-intensive, and have provided very limited sequence information on the target proteins adducted, and thus the identities of the protein targets are usually unknown. Moreover, the antibody-based methods are limited by the availability, quality, and specificity of antibodies to protein adducts, which greatly hindered the identification of specific protein targets of drugs and their clinical applications. Recently, the use of powerful MS technologies (e.g., matrix-assisted laser desorption/ionization time-of-flight) together with analytical proteomics have enabled one to separate, identify unknown protein adducts, and establish the sequence context of specific adducts by offering the opportunity to search for adducts in proteomes containing a large number of proteins with protein adducts and unmodified proteins. The present review highlights the separation and detection technologies for drug-protein adducts, with an emphasis on methodology, advantages and limitations to these techniques. Furthermore, a brief discussion of the application of these techniques to individual drugs and their target proteins will be outlined.
Collapse
Affiliation(s)
- Shufeng Zhou
- Department of Pharmacy, Faculty of Science, National University of Singapore, Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
27
|
|
28
|
Di Paolo O, de Duffard AM, Duffard R. In vivo and in vitro binding of 2,4-dichlorophenoxyacetic acid to a rat liver mitochondrial protein. Chem Biol Interact 2001; 137:229-41. [PMID: 11566291 DOI: 10.1016/s0009-2797(01)00255-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
2,4-dichlorophenoxyacetic acid (2,4-D) is a hormonal herbicide widely used in the world because of its efficacy in the control of broadleaf and woody plants. In this study we have demonstrated in vivo covalent binding of the phenoxyherbicide 2,4-D to a single protein of 52 kD (from rat liver mitochondrial preparation) detected through immunoblotting studies with the specific antiserum for 2,4-D. The direct involvement of 2,4-D in the formation of the adduct has also been demonstrated in vitro, using liver mitochondrial preparations exposed to 14C-UL-2,4-D. Radiolabeled protein separated by SDS-PAGE and afterwards electroeluted showed a single labeled protein of 52 kD. When mitochondria exposed to radiolabeled xenobiotic were devoid of their outer membrane, the specific activity observed suggest that protein involved in covalent interaction belongs to the inner mitochondrial membrane. We propose that covalent binding of the phenoxyherbicide 2,4-D to a very specific single protein of 52 kD observed in vitro and in vivo may be related to known alterations of the mitochondrial function.
Collapse
Affiliation(s)
- O Di Paolo
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531 (2000), Santa Fe, Rosario, Argentina
| | | | | |
Collapse
|
29
|
Cuff RL, Wade LT, Rychlik B, Jedlitschky GA, Burchell B. Characterisation of glucuronidation and transport in V79 cells co-expressing UGT1A1 and MRP1. Toxicol Lett 2001; 120:43-9. [PMID: 11323160 DOI: 10.1016/s0378-4274(01)00305-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The co-ordinated glucuronidation and export of compounds from cells is an important determinant in the detoxification of many compounds in vivo. Many UDP-glucuronosyltransferases (UGTs) and several multidrug resistance proteins (MRPs) have been cloned and individually expressed to assess specificity of glucuronidation and transport. However, to further understand the interplay between glucuronidation and transport we are developing stable cell lines that express different combinations of UGT and MRP isoforms. V79 cells expressing both UGT1A1 and MRP1 have been established. The ability of these cell lines to both glucuronidate and transport compounds was assessed ex vivo using estradiol and bilirubin as substrates.
Collapse
Affiliation(s)
- R L Cuff
- Department of Molecular and Cellular Pathology, Ninewells Hospital and Medical School, University of Dundee, Scotland DD1 9SY, Dundee, UK
| | | | | | | | | |
Collapse
|
30
|
Qian W, Shichi H. Cataract formation by a semiquinone metabolite of acetaminophen in mice: possible involvement of Ca(2+)and calpain activation. Exp Eye Res 2000; 71:567-74. [PMID: 11095908 DOI: 10.1006/exer.2000.0914] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acetaminophen, an analgesic/antipyretic, is metabolized by hepatic cytochrome P450 to N -acetyl- p -benzoquinone imine (NAPQI), which is transported by blood circulation to the eye and induces anterior cortical cataract in mice. In this study we injected NAPQI into the anterior chamber of mouse eye and investigated time-dependent cellular responses in the lens. After a lag period of about 2 hr following NAPQI injection, lens opacification as determined by measurement of light scattering by the lens became evident and progressively increased thereafter. There was no difference in the profile of opacity development between a P450-inducer responsive mouse strain and a non-responsive strain. During the lag period, a marked increase in free intracellular Ca(2+)in the lens epithelium was observed at 1 hr by confocal fluorescence microscopy with a Ca(2+)probe. Concurrent with the free Ca(2+)increase, there was a 300% rise in the activity of the non-lysosomal neutral protease calpain in the lens at 1 hr after NAPQI injection. Evidence indicated degradation of vimentin in the lens in which calpain activity was enhanced. Co-injection of calpain inhibitors (N-Ac-Leu-Leu-norleucinol and N-Ac-Leu-Leu-methioninal) with NAPQI protected animals completely from cataract development, although a rise in free intracellular Ca(2+)in the lens epithelium was still observed. Lenses from the protected mice did not exhibit enhanced calpain activity. These results suggest the following sequence of events as a possible mechanism of NAPQI-induced cataract. NAPQI introduced in the anterior chamber of the eye enters the lens epithelial cells and disturbs Ca(2+)homeostasis with a resultant rise in free intracellular Ca(2+)which in turn activates calpain in the epithelium. The neutral protease then degrades cellular proteins (e.g. cytoskeletal proteins) and initiates anterior cortical cataract formation.
Collapse
Affiliation(s)
- W Qian
- Kresge Eye Institute, Department of Ophthalmology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | |
Collapse
|
31
|
Lamé MW, Jones AD, Wilson DW, Dunston SK, Segall HJ. Protein targets of monocrotaline pyrrole in pulmonary artery endothelial cells. J Biol Chem 2000; 275:29091-9. [PMID: 10875930 DOI: 10.1074/jbc.m001372200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A single administration of monocrotaline to rats results in pathologic alterations in the lung and heart similar to human pulmonary hypertension. In order to produce these lesions, monocrotaline is oxidized to monocrotaline pyrrole in the liver followed by hematogenous transport to the lung where it injures pulmonary endothelium. In this study, we determined specific endothelial targets for (14)C-monocrotaline pyrrole using two-dimensional gel electrophoresis and autoradiographic detection of protein metabolite adducts. Selective labeling of specific proteins was observed. Labeled proteins were digested with trypsin, and the resulting peptides were analyzed using matrix-assisted laser desorption ionization mass spectrometry. The results were searched against sequence data bases to identify the adducted proteins. Five abundant adducted proteins were identified as galectin-1, protein-disulfide isomerase, probable protein-disulfide isomerase (ER60), beta- or gamma-cytoplasmic actin, and cytoskeletal tropomyosin (TM30-NM). With the exception of actin, the proteins identified in this study have never been identified as potential targets for pyrroles, and the majority of these proteins have either received no or minimal attention as targets for other electrophilic compounds. The known functions of these proteins are discussed in terms of their potential for explaining the pulmonary toxicity of monocrotaline.
Collapse
Affiliation(s)
- M W Lamé
- Department of Molecular Biosciences and the Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California 95616, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
The iridoid glucoside aucubin can irreversibly bind to proteins through the formation of its aglycone. In view of a possible involvement of these protein adducts in the toxicity of aucubin, we investigated the mechanism of binding of aucubin to proteins. [3H]aucubin in itself did not result in binding to protein whereas it covalently bound to rat serum albumin as a function of exposure time and dose in the presence of beta-glucosidase. The rate and extent of protein binding were significantly increased in the presence of the imine-trapping agent sodium cyanide. Oral administration of [3H]aucubin to rats showed that the total radioactivity in plasma remained at a similar level for up to 6 h once peak level was reached, suggesting that a considerable amount of radioactivity might be covalently associated with plasma proteins. The levels of radioactivity in the liver and kidney after oral dosing were higher than those after i.v. dosing. These results indicate that the open-chain aglycone of aucubin can form an imine bond with a nucleophilic site of the protein and these irreversible bindings may partially contribute to its biological and toxic effects.
Collapse
Affiliation(s)
- D H Kim
- Bioanalysis and Biotransformation Research Center, Korea Institute of Science and Technology, P.O. Box 131, Chungryang, Seoul, South Korea.
| | | | | | | |
Collapse
|
33
|
Bruno MK, Khairallah EA, Cohen SD. Inhibition of protein phosphatase activity and changes in protein phosphorylation following acetaminophen exposure in cultured mouse hepatocytes. Toxicol Appl Pharmacol 1998; 153:119-32. [PMID: 9875306 DOI: 10.1006/taap.1998.8512] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein phosphorylation was determined in cultured mouse hepatocytes exposed to an hepatotoxic concentration of acetaminophen (APAP) for selected times up to 12 h. Cultures were radiolabled with 32P-orthophosphoric acid and the cell extracts were analyzed by 2D gel electrophoresis and autoradiography. APAP exposure selectively increased the phosphorylation state of proteins of molecular weight 22, 25, 28, and 59 kDa and decreased the phosphorylation of a 26-kDa protein. Evidence is presented that these changes (1) are dependent on cytochrome P-450 activation of APAP; (2) occur well before enzyme leakage in this in vitro model; (3) are not likely attributed to GSH depletion alone; (4) are in part mimicked by okadaic acid, calyculin A, and cantharidic acid, three structurally distinct inhibitors of protein phosphatases 1 and 2A; and (5) are paralleled by a decline in protein phosphatase activity. The physiological consequences of protein phosphatase inactivation could be significant in APAP overdose since these enzymes are involved in the dephosphorylation of regulatory proteins that control many cell functions. This study also provides the first evidence for disruption in signal transduction pathways as a response to or component of APAP-induced hepatic injury.
Collapse
Affiliation(s)
- M K Bruno
- Department of Molecular and Cell Biology, University of Connecticut, Storrs 06269, USA
| | | | | |
Collapse
|
34
|
Konstandi M, Kostakis D, Johnson E, Lang MA, Marselos M. Evidence of alpha2-adrenoceptor involvement in B[alpha]P induction processes of drug-metabolizing enzymes: the effect of stress. Eur J Drug Metab Pharmacokinet 1998; 23:491-5. [PMID: 10323332 DOI: 10.1007/bf03190000] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Central to the appropriate regulation of behavioral and physiological changes induced by stress are the noradrenergic neuronal systems which have been implicated in a large number of stress-induced pathophysiological states. Endoplasmic reticulum-bound cytochromes (CYPs) play a crucial role in drug metabolism, resulting in deactivation or formation of reactive derivatives. In turn, these products may be responsible for the chemotherapeutic, mutagenic or carcinogenic properties of the parent compound. The present study assesses the effect of a specific alpha2- adrenoceptor agonist, dexmedetomidine (DEXT), on stress-induced modification of cytochrome activity in rats using a restraint stress model. The results indicated that activation of the alpha2-adrenoceptor with DEXT did not alter basal hepatic methoxyresorufin 7-dealkylase (MROD). On the other hand, it appeared to enhance MROD in benzo[alpha]pyrene (B[alpha]P) treated animals. Of interest was the finding that stress blocked DEXT-induced MROD enhancement in B[alpha]P- treated rats. In addition, DEXT had no effect on basal hepatic pentoxyresorufin 7-dealkylase (PROD), while it further enhanced the strong induction by B[alpha]P. Stress was also found to block this effect. Hepatic ethoxyresorufin 7-dealkylase (EROD) activity was strongly increased by B[alpha]P; this effect was enhanced by DEXT. In contrast, the DEXT enhanced induction was further strengthened by stress. These findings suggest that alpha2-adrenoceptors may modulate the induction of cytochromes CYP1A1, 1A2 and 2B1 by B[alpha]P in rats and that stress may modify this process. In particular, stress may regulate the inducibility of P4501A1 activity by B[alpha]P via mechanisms related to alpha2-adrenoceptors.
Collapse
Affiliation(s)
- M Konstandi
- Department of Pharmacology, Medical School, University of Ioannina, Greece
| | | | | | | | | |
Collapse
|
35
|
Konstandi M, Marselos M, Radon-Camus AM, Johnson E, Lang MA. The role of stress in the regulation of drug metabolizing enzymes in mice. Eur J Drug Metab Pharmacokinet 1998; 23:483-90. [PMID: 10323331 DOI: 10.1007/bf03189999] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The role of stress in the regulation of several enzymatic systems which are involved in the biotransformation of xenobiotics in the liver was investigated in this study using restraint stress as a stress model. The results demonstrated that stress suppressed total basal P450 content (35%) and basal ethoxyresorufin 7-dealkylase (EROD) activity (33%), while slightly increasing basal methoxyresorufin 7-dealkylase (MROD) activity (20%). Basal pentoxyresorufin 7- dealkylase (PROD) and coumarin 7-hydroxylase (COH) activities were not affected. On the other hand, restraint stress increased total P450 content in 1,4-bis[2-(3,5- dichloropyridyloxy)]benzene (TCPOBOP)-treated mice (35%), while slightly suppressing PROD activity (26%). In addition, CYP2E1 dependent p-nitrophenol hydroxylation (PNP), was suppressed (40%) by stress in TCPOBOP-treated animals and cytosolic aldehyde dehydrogenases were not affected. Although stress had no effect on basal P4502A5 activity, the inducibility of this hepatic activity increased 2-fold after stress exposure. A pronounced suppression (7-fold) in glutathione content was observed in lungs of TCPOBOP treated mice after stress, whereas basal levels remained unaffected. In addition, only a slight suppression (20%) in liver glutathione content was found in both treatment groups. Northern blot analysis revealed that restraint stress had a relatively suppressive effect on control CYP1A2 expression in the liver. In contrast, stress markedly enhanced the expression of liver CYP2A5 in TCPOBOP-treated mice, but did so to a lesser extent in controls. Stress also increased CYP2A5 mRNA in TCPOBOP-treated mice to a greater degree than the activity of the corresponding cytochrome. On the other hand, liver P4502A5 activity was found to be induced by TCPOBOP by about 2.5-fold. However, the drug does not appear to be involved in the expression of CYP2A5. Finally, although the activity of liver P4502A5 cytochrome was found to be increased 3, 8 and 27 h after stress, after which it gradually declined up to 75 h, CYP2A5 liver expression appeared to be suppressed 3, 8, 27 and 51 h after stress, while 75 h later it apparently reached normal levels. In conclusion, the results of this study showed that restraint stress significantly alters several enzymatic systems differently at a basal level than under conditions of TCPOBOP induction. In addition, stress was found to significantly interfere with the expression processes of CYP1A2 and CYP2A5.
Collapse
Affiliation(s)
- M Konstandi
- Department of Pharmacology, School of Medicine, University of Ioannina, Greece
| | | | | | | | | |
Collapse
|
36
|
Zhao C, Shichi H. Prevention of acetaminophen-induced cataract by a combination of diallyl disulfide and N-acetylcysteine. J Ocul Pharmacol Ther 1998; 14:345-55. [PMID: 9715438 DOI: 10.1089/jop.1998.14.345] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Injection of acetaminophen (APAP) (350 mg/kg body weight) into C57BL/6 mice in which cytochrome P450 (CYP) 1A1/1A2 had been induced produced acute cataract and other ocular tissue damage. Treatment of APAP-injected mice with one of the major organosulfides in garlic oil, diallyl disulfide (DADS) (200 mg/kg body weight), prevented cataract development and prolonged survival time. N-acetyl L-cysteine (NAC) (500 mg/kg body weight), a prodrug that stimulates glutathione synthesis, also prolonged survival time but was effective only weakly to prevent cataract formation. A combination of DADS and NAC completely prevented cataractogenesis, and all of the treated animals survived APAP toxicity. Neither DADS nor NAC inhibited CYP 1A1/1A2 induction as determined by their effect on the induction of hepatic microsomal ethoxyresorufin O-dealkylase (ERD) activity. However, in the in vitro enzyme assay, DADS, but not NAC, was a potent inhibitor of ERD activity (IC50 = 3.5 mM). Treatment with DADS or NAC slowed but did not stop the decrease of hepatic glutathione (GSH) content. At 4 hours after APAP injection, hepatic GSH began to increase only when DADS and NAC were administered together. These results suggest that the protective effect of DADS is due to its inhibition of biotransformation of APAP to the reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI) by CYP 1A1/1A2 enzymes and that NAC provides protection by increasing cellular cysteine level and GSH synthesis, thus facilitating detoxification of NAPQI by glutathione conjugation. Assay of plasma glutamate-pyruvate transaminase activity, an indicator of liver necrosis, showed that treatment with DADS and NAC together effectively protected the liver. Therefore, the decrease of GSH as much as 30% of normal concentration, by itself, is not responsible for liver damage. The primary cause of hepatic necrosis is rapid accumulation of NAPQI.
Collapse
Affiliation(s)
- C Zhao
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | |
Collapse
|
37
|
Qiu Y, Benet LZ, Burlingame AL. Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry. J Biol Chem 1998; 273:17940-53. [PMID: 9651401 DOI: 10.1074/jbc.273.28.17940] [Citation(s) in RCA: 224] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Liver toxicity following an overdose of acetaminophen is frequently considered a model for drug-induced hepatotoxicity. Extensive studies over many years have established that such toxicity is well correlated with liver protein arylation by acetaminophen metabolites. Identification of protein targets for covalent modifications is a challenging but necessary step in understanding how covalent binding could lead to liver toxicity. Previous approaches suffered from technical limitations, and thus over the last 10 years heroic efforts were required to determine the identity of only a few target proteins. We present a new mass spectrometry-based strategy for identification of all target proteins that now provides a comprehensive survey of the suite of liver proteins modified. After administration of radiolabeled acetaminophen to mice, the proteins in the liver tissue lysate were separated by two-dimensional polyacrylamide gel electrophoresis. In-gel digestion of the radiolabeled gel spots gave a set of tryptic peptides, which were analyzed by matrix-assisted laser desorption ionization mass spectrometry. Interrogation of data bases based on experimentally determined molecular weights of peptides and product ion tags from postsource decay mass spectra was employed for the determination of the identities of modified liver proteins. Using this method, more than 20 new drug-labeled proteins have been identified.
Collapse
Affiliation(s)
- Y Qiu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143-0446, USA
| | | | | |
Collapse
|
38
|
Shen J, Moy JA, Green MD, Guengerich FP, Baron J. Immunohistochemical demonstration of beta-naphthoflavone-inducible cytochrome P450 1A1/1A2 in rat intrahepatic biliary epithelial cells. Hepatology 1998; 27:1483-91. [PMID: 9620317 DOI: 10.1002/hep.510270604] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Although intrahepatic biliary epithelial cells are targets for certain hepatotoxic chemicals, including some procarcinogens, their ability to monooxygenate, and thereby bioactivate and inactivate xenobiotics, remains to be established. Thus, the present study was undertaken to immunohistochemically determine if cytochrome P450 (CYP) 1A1/1A2 is present and can be induced within these nonparenchymal liver cells. Immunoperoxidase and immunofluorescent staining for CYP1A1/1A2 was detected within intrahepatic biliary epithelial cells as well as hepatocytes of control rats and was markedly enhanced in both cell types by beta-naphthoflavone (BNF). Color confocal laser microscopic analyses of dual immunofluorescent staining for CYP1A1/1A2 and cytokeratins 6 and 9 (56 and 64 kd, respectively) provided unequivocal evidence for the presence and induction of CYP1A1/1A2 within intrahepatic bile duct epithelia. Moreover, microdensitometric analyses of immunoperoxidase staining intensities for CYP1A1/1A2 revealed that intrahepatic biliary epithelial cells of control rats contain 44%, 56%, and 58% as much CYP1A1/1A2 as do centrilobular, midzonal, and periportal hepatocytes, respectively. These analyses further revealed that BNF increased the content of CYP1A1/1A2 in biliary epithelial cells by approximately 120%, while CYP1A1/1A2 levels in centrilobular, midzonal, and periportal hepatocytes were increased by 82%, 159%, and 160%, respectively. The results of this study represent the first in situ demonstration that mammalian intrahepatic biliary epithelial cells contain a CYP isoform, and further that CYP1A1/1A2 can be induced in these cells by BNF. These findings therefore indicate that intrahepatic biliary epithelial cells can oxidatively metabolize xenobiotics in situ and that their ability to bioactivate and inactivate xenobiotics can be significantly enhanced by CYP1A1/1A2 induction.
Collapse
Affiliation(s)
- J Shen
- Department of Pharmacology, College of Medicine, The University of Iowa, Iowa City 52242, USA
| | | | | | | | | |
Collapse
|
39
|
Manautou JE, Silva VM, Hennig GE, Whiteley HE. Repeated dosing with the peroxisome proliferator clofibrate decreases the toxicity of model hepatotoxic agents in male mice. Toxicology 1998; 127:1-10. [PMID: 9699788 DOI: 10.1016/s0300-483x(98)00013-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pretreatment of mice with clofibrate (CFB) has been shown to protect against acetaminophen (APAP) hepatotoxicity. To determine if pretreatment with CFB prevents the toxicity of other model hepatotoxicants, male C57BL6J or CD-1 mice received 500 mg CFB/kg, i.p., daily for 10 days, and then were challenged with either 250 mg bromobenzene (BrB)/kg, 0.025 ml carbon tetrachloride (CCl4)/kg or 0.5 ml chloroform (CHCl3)/kg. Liver and kidney injury was assessed by plasma sorbitol dehydrogenase activity (SDH) and blood urea nitrogen (BUN), respectively and histopathology. Challenge with BrB significantly elevated plasma SDH activity in C57Bl6J mice. This was prevented in CFB pretreated mice receiving the same dose of BrB. Changes in BUN were not detected in either group of BrB treated mice. Similarly, pretreatment of male CD-1 mice with CFB significantly reduced CCl4-induced elevation in plasma SDH activity, with no BUN elevation detected in either group. CFB pretreatment also diminished elevation in plasma SDH activity produced by CHCl3 in CD-1 mice, while BUN was significantly elevated in both groups, indicating that CFB did not protect against CHCl3-induced nephrotoxicity. Histopathological examination of liver and kidney sections confirmed these results. This study shows that mice pretreated with CFB were protected from toxicity at 24 h after challenge with other model hepatotoxic agents besides APAP.
Collapse
Affiliation(s)
- J E Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs 06269-2092, USA.
| | | | | | | |
Collapse
|
40
|
Wang M, Dickinson RG. Hepatobiliary transport of diflunisal conjugates and taurocholate by the perfused rat liver: the effect of chronic exposure of rats to diflunisal. Life Sci 1998; 62:751-62. [PMID: 9489511 DOI: 10.1016/s0024-3205(97)01173-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acyl glucuronides are reactive electrophilic metabolites of carboxylate drugs which can form covalent adducts with endogenous macromolecules such as serum albumin and hepatic proteins. Such adducts have been suggested as initiating factors in certain immune and toxic responses to acidic drugs. In the present study, pretreatment of rats with high daily doses (50 mg/kg orally) of the non-steroidal anti-inflammatory drug (NSAID) diflunisal (DF) for 35 days, followed by perfusion of the isolated liver with 3 mg DF for 3 hr, resulted in appreciable concentrations of covalent adducts of DF with hepatic tissue (3.68 microg DF/g liver). Immunoblotting using a rabbit polyclonal DF antiserum showed the major DF-modified bands at about 110, 140 and 200 kDa. A vehicle-pretreated control group achieved adduct concentrations of only 0.37 microg DF/g liver, with the 200 kDa band not detectable in immunoblots. Elimination of DF from perfusate of the isolated perfused rat liver (IPRL) preparation was the same (t1/2 about 3.4 hr) in both DF- and vehicle-pretreated groups. Appearance of the sulfate (DS) conjugate, the major metabolite in perfusate, was also similar. However, higher concentrations of the acyl glucuronide (DAG) and phenolic glucuronide (DPG) conjugates were found in perfusate at later times, though a statistically significant difference in area under the concentration-time curve was found only in the case of DAG. At 3 hr, recoveries of dose as DAG and DPG were significantly higher in perfusate, but not in bile. No significant differences in uptake and biliary excretion of taurocholate were found between the two groups. The finding of higher perfusate concentrations of DAG and DPG could signal a minor compromise to biliary excretion processes for the glucuronides, though whether such a result is simply coincident with or attributable to DAG-derived covalent DF-protein adducts in liver remains indeterminate.
Collapse
Affiliation(s)
- M Wang
- Department of Medicine, The University of Queensland at Royal Brisbane Hospital, Australia
| | | |
Collapse
|
41
|
Zhao C, Xiong Y, Shichi H. Acetaminophen cytotoxicity in mouse eye: mitochondria in anterior tissues are the primary target. J Ocul Pharmacol Ther 1997; 13:269-76. [PMID: 9185043 DOI: 10.1089/jop.1997.13.269] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Acetaminophen (APAP) injected into C57BL/6 mice (cytochrome P450 inducer-responsive strain) that had been pretreated with b-naphthoflavone (BNF) produced ocular tissue damage, including cataract. Our previous histocytochemical studies showed that tissue damage spread in association with the flow of the aqueous humor and appeared first in the ciliary epithelium, followed by the iris and corneal endothelium and, finally, the lens. The neural retina, retinal pigmented epithelium and choroid remained unaffected. A close examination of the affected tissues indicated that mitochondria are the primary target of APAP cytotoxicity. In order to investigate whether the respiratory capacity of mitochondria is more sensitive to APAP cytotoxicity than mitochondrial morphology, we determined in this work the oxygen uptake by eye tissues dissected from BNF-pretreated and APAP-injected C57BL/6 mice. Oxygen uptake by the ciliary body/iris decreased about 60% at 90 min and 85% at 120 min after APAP administration. The oxygen uptake was inhibited about 50% by 10 microM rotenone. Since the earliest sign of mitochondrial damage was noted at 120 min, the result indicates that mitochondrial energy dysfunction precedes morphological alterations. It was also observed that oxygen uptake by the retina remained unaffected at least for 120 min after APAP administration; therefore, it is evident that the retina and, possibly, other posterior tissues as well are resistant to APAP cytotoxicity, not only in their morphology but, also, in their capacity of mitochondrial energy metabolism.
Collapse
Affiliation(s)
- C Zhao
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | |
Collapse
|
42
|
Wade LT, Kenna JG, Caldwell J. Immunochemical identification of mouse hepatic protein adducts derived from the nonsteroidal anti-inflammatory drugs diclofenac, sulindac, and ibuprofen. Chem Res Toxicol 1997; 10:546-55. [PMID: 9168252 DOI: 10.1021/tx960153t] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Reactive metabolite-modified hepatic protein adducts have been proposed to play important roles in the mechanism(s) of hepatotoxicity of nonsteroidal anti-inflammatory drugs (NSAIDs). In the present study, immunochemical techniques have been used to compare the patterns of drug-protein adducts expressed in livers of mice given single doses of one or other of three different NSAIDs. These were diclofenac and sulindac, which are widely used but potentially hepatotoxic drugs, and ibuprofen, which is considered to be nonhepatotoxic. Specific polyclonal antisera were produced by immunization of rabbits with conjugates prepared by coupling each of the NSAIDs to the carrier protein keyhole limpet hemocyanin. Immunoblotting studies revealed dose-dependent formation of major 110 kDa polypeptide adducts in livers from mice sacrificed 6 h after administration of single doses of either diclofenac (0-300 mg/kg) or sulindac (0-100 mg/kg). Lower levels of several other adducts, of 140 and 200 kDa, were also expressed in livers from these animals. In contrast, livers from mice treated with ibuprofen (0-200 mg/kg) predominantly expressed a 60 kDa adduct and only relatively low levels of a 110 kDa adduct. The various adducts were shown by differential centrifugation to be concentrated in the nuclear fraction of liver homogenates. Those derived from diclofenac and sulindac were further localized, by Percoll density gradient centrifugation, to a subfraction which contained a high activity of the bile canalicular marker enzyme alkaline phosphatase. This suggests that they are concentrated in the bile canalicular domain of hepatocytes. The different patterns of adduct formation raise the possibility that formation of certain NSAID protein adducts, particularly 110 kDa adducts, has toxicological significance.
Collapse
Affiliation(s)
- L T Wade
- Pharmacology and Toxicology, Imperial College School of Medicine at St. Mary's, London, U.K
| | | | | |
Collapse
|
43
|
Abstract
1. Pharmacokinetics aids interpretation of the dose-response relationship in individual toxicology studies. 2. When used to compare across studies, even in a single species other factors, including variation in pharmacodynamic response, must be taken into account. Variation in pharmacodynamic response becomes more profound when one compares across species. 3. Examples do occur where plasma concentration-response relationships are constant across species, particularly when corrected for unbound drug. These examples should not be taken as support, however, of a general universal principle. 4. Owing to multiple factors such as species differences in receptors, enzymes and ion channels, dose or plasma concentration-response relationships can vary enormously across species. In the light of this, the results of toxicology studies should be viewed as qualitative rather than quantitative. Once sufficient clinical experience is gained the human database is the overriding measure of drug safety.
Collapse
Affiliation(s)
- D A Smith
- Department of Drug Metabolism, Pfizer Central Research, Sandwich, UK
| |
Collapse
|
44
|
Cohen SD, Pumford NR, Khairallah EA, Boekelheide K, Pohl LR, Amouzadeh HR, Hinson JA. Selective protein covalent binding and target organ toxicity. Toxicol Appl Pharmacol 1997; 143:1-12. [PMID: 9073586 DOI: 10.1006/taap.1996.8074] [Citation(s) in RCA: 239] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Protein covalent binding by xenobiotic metabolites has long been associated with target organ toxicity but mechanistic involvement of such binding has not been widely demonstrated. Modern biochemical, molecular, and immunochemical approaches have facilitated identification of specific protein targets of xenobiotic covalent binding. Such studies have revealed that protein covalent binding is not random, but rather selective with respect to the proteins targeted. Selective binding to specific cellular target proteins may better correlate with toxicity than total protein covalent binding. Current research is directed at characterizing and identifying the targeted proteins and clarifying the effect of such binding on their structure, function, and potential roles in target organ toxicity. The approaches employed to detect and identify the tartgeted proteins are described. Metabolites of acetaminophen, halothane, and 2,5-hexanedione form covalently bound adducts to recently identified protein targets. The selective binding may influence homeostatic or other cellular responses which in turn contribute to drug toxicity, hypersensitivity, or autoimmunity.
Collapse
Affiliation(s)
- S D Cohen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs 06269-2092, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
More than 20 years have passed since the early reports of acute hepatotoxicity with APAP overdose. During that period investigative research to discover the "mechanism" underlying the toxicity has been conducted in many species and strains of intact animals as well as in a variety of in vitro and culture systems. Such work has clarified the primary role of biotransformation and the protective role of GSH. Understanding the former provides explanations for the toxic interactions which may occur with alcohol or other xenobiotics, while understanding of the latter led to the development of antidotes for the treatment of acute poisoning. Acetaminophen (APAP)-induced hepatotoxicity: roles for protein arylation. Initiating events in toxicity require biotransformation of APAP to NAPQI followed by arylation of several important proteins with subsequent alteration of protein structure and function. The immediate consequence of the alterations is detectable in several organelles and these may represent multiple initiating events which are depicted as acting in concert to cause cell injury (large arrowheads). Arylation of cytosolic 58-ABP with subsequent translocation to the nucleus is depicted as a possible signaling mechanism for determining outcome at the cell or organ level (within dotted boundary). For simplicity NAPQI's potentials for oxidizing protein sulfhydryls and direct binding to DNA have been omitted. Significant light has also been shed on the biochemical and cellular events which accompany APAP-induced hepatotoxicity. However, such studies have not identified a unique mechanism of toxicity that is universally accepted. The recent identification of several protein targets which become arylated during toxicity--along with the findings that arylation of some of those target proteins results in loss of protein function--demonstrates that covalent binding does, indeed, have biological consequences and is not merely an indicator of the fleeting presence of reactive electrophiles. These observations further suggest that multiple independent insults to the cell may be involved in toxicity. it is now apparent that the concept of a multistage process that involves both initiation and progression events is appropriate for APAP toxicity, and it is unlikely that a unique initiating event will ever be identified. In light of recent findings it is more likely that a number of such cellular events occur very early after toxic overdosage, and that they collectively set in motion and perpetuate the biochemical, cellular, and molecular processes which will determine outcome. The importance of 58-ABP arylation with early, apparently selective, translocation to the nucleus remains to be elucidated. To date there is nothing to suggest that this represents an initiating event in toxicity. rather it is plausible that the translocation may play a role in signaling electrophile presence and in calling for cellular defense against electrophile insult. This is reflected in the hypothetical model presented in Fig. 3. Critical experimental testing of this model will advance our understanding of the cellular and molecular responses to toxic electrophile insult.
Collapse
Affiliation(s)
- S D Cohen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs 06269, USA
| | | |
Collapse
|
46
|
Castell JV, Gómez-Lechón MJ, Ponsoda X, Bort R. In vitro investigation of the molecular mechanisms of hepatotoxicity. ARCHIVES OF TOXICOLOGY. SUPPLEMENT. = ARCHIV FUR TOXIKOLOGIE. SUPPLEMENT 1997; 19:313-21. [PMID: 9079218 DOI: 10.1007/978-3-642-60682-3_29] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- J V Castell
- Unidad de Hepatología Experimental, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | |
Collapse
|
47
|
Abstract
Many xenobiotics are metabolically activated to electrophilic intermediates that form covalent adducts with proteins; the mechanism of toxicity is either intrinsic or idiosyncratic in nature. Many intrinsic toxins covalently modify cellular proteins and somehow initiate a sequence of events that leads to toxicity. Major protein adducts of several intrinsic toxins have been identified and demonstrate significant decreases in enzymatic activity. The reactivity of intermediates and subcellular localization of major targets may be important in the toxicity. Idiosyncratic toxicities are mediated through either a metabolic or immune-mediated mechanism. Xenobiotics that cause hypersensitivity/autoimmunity appear to have a limited number of protein targets, which are localized within the subcellular fraction where the electrophile is produced, are highly substituted, and are accessible to the immune system. Metabolic idiosyncratic toxins appear to have limited targets and are localized within a specific subcellular fraction. Identification of protein targets has given us insights into mechanisms of xenobiotic toxicity.
Collapse
Affiliation(s)
- N R Pumford
- Division of Toxicology, University of Arkansas for Medical Sciences, Little Rock 72205-7199, USA
| | | |
Collapse
|
48
|
Abstract
Despite considerable progress in the understanding of the mechanism of liver toxicity we are not yet able to design non-hepatotoxic molecules rationally. Also, there is no "universal" in vitro primary screening approach for early identification of hepatotoxic molecules. In most cases hepatotoxicity is detected at later stages of drug development in animal toxicity studies or clinical trials. Although the liver is the most common target organ for drug candidates in animal toxicity studies, hepatotoxicity rarely leads to cessation of drug development during the preclinical phase. Indeed, contrary to other target organs, liver toxicity is usually reversible and can be monitored in man by sensitive serum enzyme tests. Therefore in many cases a compound found hepatotoxic in an animal species will be tested in man for a definitive assessment of its hepatotoxic potential. Liver toxicity in man may be acceptable when a drug has major therapeutic potential. In this situation mechanistic studies are essential to assess the risk in man and in some cases to identify protective agents. When liver toxicity leads to project termination a secondary screening approach may be envisaged if biologically active analogs are available.
Collapse
Affiliation(s)
- F Ballet
- Rhône-Poulenc Rorer, Drug Safety Division, Vitry-sur-Seine, France
| |
Collapse
|
49
|
Identification of a 54-kDa mitochondrial acetaminophen-binding protein as aldehyde dehydrogenase. Toxicol Appl Pharmacol 1996. [DOI: 10.1016/s0041-008x(96)80036-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
de Zwart LL, Hermanns RC, Meerman JH, Commandeur JN, Vermeulen NP. Disposition in rat of [2-3H]-trans-4-hydroxy-2,3-nonenal, a product of lipid peroxidation. Xenobiotica 1996; 26:1087-100. [PMID: 8905921 DOI: 10.3109/00498259609167424] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
1. 4-Hydroxy-2,3-nonenal (HNE) is an end product of lipid peroxidation (LPO) and a well known cytotoxic aldehyde that exhibits a variety of biological effects. In this study the in vivo disposition and covalent binding of i.p. administered [2-3H]HNE was examined in the rat. 2. It was found that several metabolites of [2-3H]HNE are excreted in urine among which at least four mercapturic acids. 1,4-Dihydroxynonane mercapturic acid (DHN-MA) appeared to be the most abundant mercapturic acid excreted in urine (3.5% of the dose) and the excretion of the other three mercapturic acids amounted to 2% of the dose. 3. Within 48 h following i.p. administration of 5 or 25 mumol/kg bodyweight [2-3H]HNE (specific activity 4 microCi/mumol) about 25% of the radioactivity was excreted in urine, whereas 18% of the radioactivity appeared in the faeces. 4. After 48 h, 7% of the radioactivity was still present in the liver and 0.2% in other organs, but this radioactivity appeared to not to be covalently bound to cellular macromolecules. It was found that only 0.13% of the radioactivity was covalently bound in the liver and even less in other organs.
Collapse
Affiliation(s)
- L L de Zwart
- Leiden/Amsterdam Center for Drug Research, Free University, Department of Pharmacochemistry, The Netherlands
| | | | | | | | | |
Collapse
|