1
|
Wang W, Wang SK, Wang Q, Zhang Z, Li B, Zhou ZD, Zhang JF, Lin C, Chen TX, Jin Z, Tang YZ. Diclofenac and eugenol hybrid with enhanced anti-inflammatory activity through activating HO-1 and inhibiting NF-κB pathway in vitro and in vivo. Eur J Med Chem 2023; 259:115669. [PMID: 37517204 DOI: 10.1016/j.ejmech.2023.115669] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023]
Abstract
A series of diclofenac hybrid molecules were synthesized and evaluated for their NO-inhibitory ability in LPS-induced RAW 264.7 macrophage cells. Among them, compound 1 showed the highest NO-inhibitory ability (approximately 66%) and no significant cytotoxicity. Compound 1 exhibited superior NF-κB-inhibitory ability compared to diclofenac through the activation of Nrf2/HO-1 signaling pathway in RAW 264.7. 20 mg/kg compound 1 resulted in remarkable colitis improvement in dextran sulfate sodium (DSS)-induced mice model by up-regulating HO-1 and down-regulating phosphorylation level of NF-κB p65. Moreover, 50 mg/kg dose of compound 1 showed a lower ulcerogenic potential compared to diclofenac in rats. The diclofenac-eugenol hybrid (compound 1) may serve as a novel anti-inflammatory agent based on its role in inhibiting the NF-κB signaling pathway and activating HO-1 expression with no toxicity in vitro and in vivo.
Collapse
Affiliation(s)
- Wei Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Shou-Kai Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhe Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Bo Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zi-Dan Zhou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jian-Feng Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Chao Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Ting-Xiao Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
2
|
Badr H, Kozai D, Sakaguchi R, Numata T, Mori Y. Different Contribution of Redox-Sensitive Transient Receptor Potential Channels to Acetaminophen-Induced Death of Human Hepatoma Cell Line. Front Pharmacol 2016; 7:19. [PMID: 26903865 PMCID: PMC4746322 DOI: 10.3389/fphar.2016.00019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/21/2016] [Indexed: 01/30/2023] Open
Abstract
Acetaminophen (APAP) is a safe analgesic antipyretic drug at prescribed doses. Its overdose, however, can cause life-threatening liver damage. Though, involvement of oxidative stress is widely acknowledged in APAP-induced hepatocellular death, the mechanism of this increased oxidative stress and the associated alterations in Ca2+ homeostasis are still unclear. Among members of transient receptor potential (TRP) channels activated in response to oxidative stress, we here identify that redox-sensitive TRPV1, TRPC1, TRPM2, and TRPM7 channels underlie Ca2+ entry and downstream cellular damages induced by APAP in human hepatoma (HepG2) cells. Our data indicate that APAP treatment of HepG2 cells resulted in increased reactive oxygen species (ROS) production, glutathione (GSH) depletion, and Ca2+ entry leading to increased apoptotic cell death. These responses were significantly suppressed by pretreatment with the ROS scavengers N-acetyl-L-cysteine (NAC) and 4,5-dihydroxy-1,3-benzene disulfonic acid disodium salt monohydrate (Tiron), and also by preincubation of cells with the glutathione inducer Dimethylfumarate (DMF). TRP subtype-targeted pharmacological blockers and siRNAs strategy revealed that suppression of either TRPV1, TRPC1, TRPM2, or TRPM7 reduced APAP-induced ROS formation, Ca2+ influx, and cell death; the effects of suppression of TRPV1 or TRPC1, known to be activated by oxidative cysteine modifications, were stronger than those of TRPM2 or TRPM7. Interestingly, TRPV1 and TRPC1 were labeled by the cysteine-selective modification reagent, 5,5′-dithiobis (2-nitrobenzoic acid)-2biotin (DTNB-2Bio), and this was attenuated by pretreatment with APAP, suggesting that APAP and/or its oxidized metabolites act directly on the modification target cysteine residues of TRPV1 and TRPC1 proteins. In human liver tissue, TRPV1, TRPC1, TRPM2, and TRPM7 channels transcripts were localized mainly to hepatocytes and Kupffer cells. Our findings strongly suggest that APAP-induced Ca2+ entry and subsequent hepatocellular death are regulated by multiple redox-activated cation channels, among which TRPV1 and TRPC1 play a prominent role.
Collapse
Affiliation(s)
- Heba Badr
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | - Daisuke Kozai
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | - Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto UniversityKyoto, Japan
| | - Tomohiro Numata
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto UniversityKyoto, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyoto, Japan; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto UniversityKyoto, Japan; Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto UniversityKyoto, Japan
| |
Collapse
|
3
|
Leth R, Rydberg P, Jørgensen FS, Olsen L. Density Functional Theory Study on the Formation of Reactive Benzoquinone Imines by Hydrogen Abstraction. J Chem Inf Model 2015; 55:660-6. [DOI: 10.1021/ci500653b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Rasmus Leth
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Patrik Rydberg
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Lars Olsen
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
4
|
Wang AY, Lian LH, Jiang YZ, Wu YL, Nan JX. Gentiana manshurica Kitagawa prevents acetaminophen-induced acute hepatic injury in mice via inhibiting JNK/ERK MAPK pathway. World J Gastroenterol 2010; 16:384-91. [PMID: 20082487 PMCID: PMC2807962 DOI: 10.3748/wjg.v16.i3.384] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the in vivo hepatoprotective effects and mechanisms of Gentiana manshurica Kitagawa (GM) in acetaminophen (APAP)-induced liver injury in mice.
METHODS: GM (200, 150 or 50 mg/kg body weight) or N-acetyl-L-cysteine (NAC; 300 mg/kg body weight) was administrated orally with a single dose 2 h prior to APAP (300 mg/kg body weight) injection in mice.
RESULTS: APAP treatment significantly depleted hepatic glutathione (GSH), increased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) and malonyldialdehyde (MDA) and 4-hydroxynonenal levels, and decreased hepatic activity of glutathione peroxidase (GSH-px) and superoxide dismutase (SOD). However, the pretreatment of GM significantly alleviated APAP-induced oxidative stress by increasing GSH content, decreasing serum ALT, AST and MDA, and retaining the activity of GSH-px and SOD in the liver. Furthermore, GM pretreatment can inhibit caspase-3 activation and phosphorylation of c-Jun-NH2-terminal protein kinase 2 (JNK1/2) and extracellular signal-regulated kinase (ERK). GM also remarkably attenuated hepatocyte apoptosis confirmed by the terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling method.
CONCLUSION: Hepatoprotective effects of GM against APAP-induced acute toxicity are mediated either by preventing the decline of hepatic antioxidant status or its direct anti-apoptosis capacity. These results support that GM is a potent hepatoprotective agent.
Collapse
|
5
|
Nishida T, Matsura T, Nakada J, Togawa A, Kai M, Sumioka I, Minami Y, Inagaki Y, Ishibe Y, Ito H, Ohta Y, Yamada K. Geranylgeranylacetone protects against acetaminophen-induced hepatotoxicity by inducing heat shock protein 70. Toxicology 2006; 219:187-96. [PMID: 16377054 DOI: 10.1016/j.tox.2005.11.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 11/15/2005] [Accepted: 11/15/2005] [Indexed: 11/20/2022]
Abstract
Geranylgeranylacetone (GGA), an anti-ulcer drug, has been reported to induce heat shock protein (HSP) 70 in several animal organs. The present study was performed to determine whether GGA protects mouse liver against acetaminophen (APAP)-induced injury and whether it has potential as a therapeutic agent for APAP overdose. Hepatic damage was induced by single oral administration of APAP (500 mg/kg). GGA at 400 mg/kg was given orally 4 or 8h before, or 0.5h after APAP administration. Treatment of mice with GGA 4h before or 0.5h after APAP administration suppressed increases in transaminase activities and ammonia content in blood as well as hepatic necrosis. Such GGA treatment significantly increased hepatic HSP70 accumulation after APAP administration. Furthermore, GGA inhibited increases in hepatic lipid peroxide content and hepatic myeloperoxidase activity after APAP administration. In contrast, GGA neither inhibited hepatic cytochrome P450 2E1 activity nor suppressed hepatic glutathione depletion after APAP administration. The protective effect of GGA treatment 4h before APAP on hepatotoxicity induced by APAP was completely inhibited with quercetin, known as an HSP inhibitor. In conclusion, GGA has been identified as a new antidote to APAP injury, acting by induction of HSP70. The potential of GGA as a therapeutic tool is strongly supported by its ability to inhibit hepatic injury even when administered after ingestion of APAP.
Collapse
Affiliation(s)
- Tadashi Nishida
- Division of Medical Biochemistry, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
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
|
7
|
Ataie G, Safarian S, Divsalar A, Saboury AA, Moosavi-Movahedi AA, Ranjbar B, Cristalli G, Mardanian S. Kinetic and structural analysis of the inhibition of adenosine deaminase by acetaminophen. J Enzyme Inhib Med Chem 2004; 19:71-8. [PMID: 15202496 DOI: 10.1080/14756360310001632741] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Kinetic and thermodynamic studies have been made on the effect of acetaminophen on the activity and structure of adenosine deaminase in 50 mM sodium phosphate buffer pH 7.5, at two temperatures of 27 and 37 degrees C using UV spectrophotometry, circular dichroism (CD) and fluorescence spectroscopy. Acetaminophen acts as a competitive inhibitor at 27 degrees C (Ki = 126 microM) and an uncompetitive inhibitor at 37 degrees C (Ki = 214 microM). Circular dichroism studies do not show any considerable effect on the secondary structure of adenosine deaminase by increasing the temperature from 27 to 37 degrees C. However, the secondary structure of the protein becomes more compact at 37 degrees C in the presence of acetaminophen. Fluorescence spectroscopy studies show considerable change in the tertiary structure of the protein by increasing the temperature from 27 to 37 degrees C. Also, the fluorescence spectrum of the protein incubated with different concentrations of acetaminophen show different inhibition behaviors by the effector at the two temperatures.
Collapse
Affiliation(s)
- G Ataie
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
8
|
|
9
|
Abstract
The analgesic acetaminophen causes a potentially fatal, hepatic centrilobular necrosis when taken in overdose. The initial phases of toxicity were described in Dr. Gillette's laboratory in the 1970s. These findings indicated that acetaminophen was metabolically activated by cytochrome P450 enzymes to a reactive metabolite that depleted glutathione (GSH) and covalently bound to protein. It was shown that repletion of GSH prevented the toxicity. This finding led to the development of the currently used antidote N-acetylcysteine. The reactive metabolite was subsequently identified to be N-acetyl-p-benzoquinone imine (NAPQI). Although covalent binding has been shown to be an excellent correlate of toxicity, a number of other events have been shown to occur and are likely important in the initiation and repair of toxicity. Recent data have shown that nitrated tyrosine residues as well as acetaminophen adducts occur in the necrotic cells following toxic doses of acetaminophen. Nitrotyrosine was postulated to be mediated by peroxynitrite, a reactive nitrogen species formed by the very rapid reaction of superoxide and nitric oxide (NO). Peroxynitrite is normally detoxified by GSH, which is depleted in acetaminophen toxicity. NO synthesis (serum nitrate plus nitrite) was dramatically increased following acetaminophen. In inducible nitric oxide synthase (iNOS) knockout mice, acetaminophen did not increase NO synthesis or tyrosine nitration; however, histological evidence indicated no difference in toxicity. Acetaminophen did not cause hepatic lipid peroxidation in wild-type mice but did cause lipid peroxidation in iNOS knockout mice. These data suggest that NO may play a role in controlling lipid peroxidation and that reactive nitrogen/oxygen species may be important in toxicity. The source of the superoxide has not been identified, but our recent finding that NADPH oxidase knockout mice were equally sensitive to acetaminophen and had equal nitration of tyrosine suggests that the superoxide is not from the activation of Kupffer cells. It was postulated that NAPQI-mediated mitochondrial injury may be the source of the superoxide. In addition, the significance of cytokines and chemokines in the development of toxicity and repair processes has been demonstrated by several recent studies. IL-1beta is increased early in acetaminophen toxicity and may be important in iNOS induction. Other cytokines, such as IL-10, macrophage inhibitory protein-2 (MIP-2), and monocyte chemoattractant protein-1 (MCP-1), appear to be involved in hepatocyte repair and the regulation of proinflammatory cytokines.
Collapse
Affiliation(s)
- Laura P James
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | | | | |
Collapse
|
10
|
Valero E, Varón R, García-Carmona F. Tyrosinase-mediated oxidation of acetaminophen to 4-acetamido-o-benzoquinone. Biol Chem 2002; 383:1931-9. [PMID: 12553730 DOI: 10.1515/bc.2002.217] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Based on its monophenolic structure and given its pharmacological and toxicological importance, the ability of tyrosinase to oxidize acetaminophen was studied for the first time. Progress curves showed a transient phase characteristic of the monophenolase activity of tyrosinase prior to attaining the steady-state. The duration of this transient phase strongly increased with the drug concentration, which would partly explain why paracetamol oxidation by tyrosinase has not been studied hitherto. The pathway is enhanced by the presence of minute amounts of L-dopa, which shortens the length of the lag period. Acetaminophen oxidation was inhibited by tropolone, a selective inhibitor of tyrosinase. The presence of the corresponding o-diphenol as intermediate was demonstrated with ascorbic acid by chemical oxidation using NaIO4 and by HPLC analysis, indicating that acetaminophen is oxidized by the monophenolase activity of tyrosinase to its corresponding o-quinone. These results contribute to our knowledge of the oxidation mechanisms of acetaminophen.
Collapse
Affiliation(s)
- Edelmira Valero
- Departamento de Química-Fisica, Escuela Politécnica Superior de Albacete, Universidad de Castilla-La Mancha, Campus Universitario, E-02071 Albacete, Spain
| | | | | |
Collapse
|
11
|
Bessems JG, Vermeulen NP. Paracetamol (acetaminophen)-induced toxicity: molecular and biochemical mechanisms, analogues and protective approaches. Crit Rev Toxicol 2001; 31:55-138. [PMID: 11215692 DOI: 10.1080/20014091111677] [Citation(s) in RCA: 455] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An overview is presented on the molecular aspects of toxicity due to paracetamol (acetaminophen) and structural analogues. The emphasis is on four main topics, that is, bioactivation, detoxication, chemoprevention, and chemoprotection. In addition, some pharmacological and clinical aspects are discussed briefly. A general introduction is presented on the biokinetics, biotransformation, and structural modification of paracetamol. Phase II biotransformation in relation to marked species differences and interorgan transport of metabolites are described in detail, as are bioactivation by cytochrome P450 and peroxidases, two important phase I enzyme families. Hepatotoxicity is described in depth, as it is the most frequent clinical observation after paracetamol-intoxication. In this context, covalent protein binding and oxidative stress are two important initial (Stage I) events highlighted. In addition, the more recently reported nuclear effects are discussed as well as secondary events (Stage II) that spread over the whole liver and may be relevant targets for clinical treatment. The second most frequent clinical observation, renal toxicity, is described with respect to the involvement of prostaglandin synthase, N-deacetylase, cytochrome P450 and glutathione S-transferase. Lastly, mechanism-based developments of chemoprotective agents and progress in the development of structural analogues with an improved therapeutic index are outlined.
Collapse
Affiliation(s)
- J G Bessems
- Leiden/Amsterdam Center for Drug Research, Department of Pharmacochemistry, Vrije Universiteit, Amsterdam, The Netherlands
| | | |
Collapse
|
12
|
Huang R, Okuno H, Takasu M, Takeda S, Kano H, Shiozaki Y, Inoue K. Effects of rifampin on the glutathione depletion and cytochrome c reduction by acetaminophen reactive metabolites in an in vitro P450 enzyme system. JAPANESE JOURNAL OF PHARMACOLOGY 2000; 83:182-90. [PMID: 10952066 DOI: 10.1254/jjp.83.182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study examined whether rifampin attenuated glutathione (GSH) depletion by acetaminophen reactive metabolites generated in the in vitro P450 enzyme system prepared from mouse liver and the possible mechanism involved in this effect. The results showed that GSH concentration was decreased concentration-dependently by acetaminophen in the in vitro P450 enzyme system. Rifampin significantly attenuated acetaminophen-mediated GSH depletion in a concentration-dependent manner. The concentration-response curve for GSH depletion of acetaminophen was shifted to the right in a parallel fashion in the presence of rifampin at the concentration of 3.2 x 10(-5) M, which appeared to result from the competitive binding of rifampin to acetaminophen metabolites. Cytochrome c was markedly reduced by acetaminophen metabolites in this enzyme system, and GSH concentration-dependently increased the cytochrome c reduction by acetaminophen metabolites. These findings suggested that cytochrome c was reduced by the GSH conjugate of acetaminophen metabolites rather than by acetaminophen-derived superoxide anion (O2*-) and other unbound free radicals. Rifampin was shown to possess an effect similar to that of GSH. It is concluded that the decrease in GSH depletion by rifampin is most likely attributable to the binding of rifampin to the acetaminophen toxic species, and the increase in cytochrome c reduction by rifampin is attributable to the conjugate formed between rifampin and acetaminophen metabolites.
Collapse
Affiliation(s)
- R Huang
- Third Department of Internal Medicine, Kansai Medical University, Moriguchi, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Bessems JG, de Groot MJ, Baede EJ, te Koppele JM, Vermeulen NP. Hydrogen atom abstraction of 3,5-disubstituted analogues of paracetamol by horseradish peroxidase and cytochrome P450. Xenobiotica 1998; 28:855-75. [PMID: 9764928 DOI: 10.1080/004982598239100] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
1. The formation of free radicals during enzyme catalysed oxidation of eight 3,5-disubstituted analogues of paracetamol (PAR) has been studied. A simple peroxidase system as well as cytochrome P450-containing systems were used. Radicals were detected by electron spin resonance (ESR) on incubation of PAR and 3,5-diCH3-, 3,5-diC2H5-, 3,5-ditC4H9-, 3,5-diOCH3-, 3,5-diSCH3-, 3,5-diF-, 3,5-diCl- and 3,5-diBr-substituted analogues of PAR with horseradish peroxidase in the presence of hydrogen peroxide (H2O2). Initial analysis of the observed ESR spectra revealed all radical species to be phenoxy radicals, based on the absence of dominant nitrogen hyperfine splittings. No radicals were detected in rat liver cytochrome P450-containing microsomal or reconstituted systems. 2. To rationalize the observed ESR spectra, hydrogen atom abstraction of PAR and four of the 3,5-disubstituted analogues (3,5-diCH3-, 3,5-diOCH3-, 3,5-diF- and 3,5-diCl-PAR) was calculated using ab initio calculations, and a singlet oxygen atom was used as the oxidizing species. The calculations indicated that for all compounds studied an initial hydrogen atom abstraction from the phenolic hydroxyl group is favoured by approximately 125 kJ/mol over an initial hydrogen atom abstraction from the acetylamino nitrogen atom, and that after hydrogen abstraction from the phenolic hydroxyl group, the unpaired electron remains predominantly localised at the phenoxy oxygen atom (+/-85%). 3. The experimental finding of phenoxy radicals in horseradish peroxidase/H2O2 incubations paralleled these theoretical findings. The failure to detect experimentally phenoxy radicals in cytochrome P450-catalysed oxidation of any of the eight 3,5-disubstituted PAR analogues is more likely due to the reducing effects that agents like NADPH and protein thiol groups have on phenoxy radicals rather than on the physical instability of the respective substrate radicals.
Collapse
Affiliation(s)
- J G Bessems
- Leiden/Amsterdam Center for Drug Research, Department of Pharmacochemistry, Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
14
|
Jones AL. Mechanism of action and value of N-acetylcysteine in the treatment of early and late acetaminophen poisoning: a critical review. JOURNAL OF TOXICOLOGY. CLINICAL TOXICOLOGY 1998; 36:277-85. [PMID: 9711192 DOI: 10.3109/15563659809028022] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The mechanism of action of N-acetylcysteine in early acetaminophen poisoning is well understood, but much remains to be learned of the mechanism of its possible benefit in acetaminophen poisoning presenting beyond 15 hours. METHODS Selective review of medical literature. N-acetylcysteine should be used in all cases of early acetaminophen poisoning where the plasma acetaminophen concentration lies "above the line;" which line is chosen depends on individual preference and whether enzyme induction is suspected. Particular care should be taken with the use of the nomogram for patients with chronic excess ingestion of acetaminophen or for those who have taken slow-release formulations. CONCLUSIONS While there is a trend suggesting a beneficial effect of N-acetylcysteine in some patients presenting beyond 15 hours, further research is necessary to establish just how effective N-acetylcysteine is, particularly in patients presenting with fulminant hepatic failure. Candidate mechanisms for a beneficial effect in-clude improvement of liver blood flow, glutathione replenishment, modification of cytokine production, and free radical or oxygen scavenging. Hemody-namic and oxygen delivery and utilization parameters must be monitored carefully during delayed N-acetylcysteine treatment of patients with fulminant hepatic failure, as unwanted vasodilation may be deleterious to the maintenance of mean arterial blood pressure.
Collapse
Affiliation(s)
- A L Jones
- Scottish Poisons Information Bureau, Royal Infirmary of Edinburgh, Scotland.
| |
Collapse
|
15
|
Weis M, Rundgren M, Nelson S, Moldéus P. Peroxidase-catalyzed oxidation of 3,5-dimethyl acetaminophen causes cell death by selective protein thiol modification in isolated rat hepatocytes. Chem Biol Interact 1996; 100:255-65. [PMID: 8653807 DOI: 10.1016/0009-2797(96)03703-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In this study we used a peroxidase model system (glucose/glucose oxidase and horseradish peroxidase) to investigate the effect of extracellularly generated reactive metabolites of 3,5-Me2-acetaminophen on cell viability and on cellular thiol levels. Incubation of hepatocytes with 3,5-Me2-acetaminophen in the presence of glucose/glucose oxidase and horseradish peroxidase caused a concentration-dependent loss of cell viability. Loss of viability was associated with decreased protein thiol levels. Addition of the reducing agent DTT, but not catalase, during the incubation restored cellular protein thiol levels and arrested the cell killing. Protein thiol depletion occurred selectively to the mitochondrial and microsomal fractions and was specific for a very limited number of protein bands. The data suggest that the oxidative modification of individual protein cysteine residues within the latter two organelle fractions is critically involved in the mechanism of toxicity.
Collapse
Affiliation(s)
- M Weis
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institute, Stockholm, Sweden
| | | | | | | |
Collapse
|
16
|
Tingle MD, Jewell H, Maggs JL, O'Neill PM, Park BK. The bioactivation of amodiaquine by human polymorphonuclear leucocytes in vitro: chemical mechanisms and the effects of fluorine substitution. Biochem Pharmacol 1995; 50:1113-9. [PMID: 7575670 DOI: 10.1016/0006-2952(95)00236-s] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Amodiaquine, a 4-aminoquinoline antimalarial, has been associated with hepatitis and agranulocytosis in humans. Drug hypersensitivity reactions, especially agranulocytosis, have been attributed to reactive intermediates generated by the oxidants discharged from stimulated polymorphonuclear leucocytes (PMN). The metabolism of amodiaquine to both stable and chemically reactive metabolites by human PMN has been investigated in vitro. Incubation of [14C]-amodiaquine with PMN resulted in irreversible binding of radiolabel to protein and depletion of intracellular reduced glutathione, which were enhanced by phorbol myristate acetate (PMA), a PMN activator. Two metabolites were identified: the C-5' glutathione adduct of amodiaquine, derived from both endogenous and exogenous glutathione, and 4-amino-7-chloroquinoline, which was presumed to be formed by hydrolysis of amodiaquine quinoneimine. Desethylamodiaquine, the major plasma metabolite of amodiaquine in humans, also underwent bioactivation to a chemically reactive species in the presence of PMA-stimulated PMN. Substitution of the 4'-hydroxyl group in amodiaquine with fluorine significantly reduced irreversible binding to protein and abolished depletion of intracellular glutathione in the presence of PMA. These findings indicate that the bioactivation of amodiaquine by PMN is associated with the formation of a quinoneimine intermediate. Such a reactive metabolite, if produced in PMN or bone marrow in vivo, may be responsible for the drug's myelotoxicity.
Collapse
Affiliation(s)
- M D Tingle
- Department of Pharmacology and Therapeutics, University of Liverpool, U.K
| | | | | | | | | |
Collapse
|
17
|
Amimoto T, Matsura T, Koyama SY, Nakanishi T, Yamada K, Kajiyama G. Acetaminophen-induced hepatic injury in mice: the role of lipid peroxidation and effects of pretreatment with coenzyme Q10 and alpha-tocopherol. Free Radic Biol Med 1995; 19:169-76. [PMID: 7649488 DOI: 10.1016/0891-5849(94)00233-a] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
This study was performed to determine whether oxidative stress contributed to the initiation or progression of hepatic injury produced by acetaminophen (APAP). Treatment of fasted mice with APAP (400 mg/kg, I.P.) led to hepatic injury as indicated by a marked elevation of plasma alanine aminotransferase (ALT). APAP caused an increased amount of thiobarbituric acid-reactive substance (TBARS), which was accompanied by a loss of reduced forms of coenzyme Q9 (CoQ9H2) and coenzyme Q10 (CoQ10H2) functioning as antioxidants. APAP also markedly decreased hepatic reduced glutathione (GSH) levels. Pretreatment with CoQ10 (5 mg/kg, I.V.) reduced hepatic TBARS levels to 30% and plasma ALT levels to 26% of placebo pretreatment levels without affecting hepatic GSH levels at 3 h of APAP treatment. alpha-Tocopherol (alpha-Toc) (20 mg/kg, I.V.) pretreatment also reduced hepatic TBARS levels to 13% and plasma ALT levels to 27% of placebo pretreatment levels without affecting hepatic GSH levels. These results suggest that oxidative stress followed by lipid peroxidation might play a role in the pathogenesis of APAP-induced hepatic injury, and pretreatment with lipid-soluble antioxidants such as CoQ10 and alpha-Toc can limit hepatic injury produced by APAP.
Collapse
Affiliation(s)
- T Amimoto
- Department of Internal Medicine, Hiroshima University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Oxygen free radicals are very reactive molecules which can react with every cellular component. They are normally produced in organisms being involved in various biologic reactions. However, too high levels of these partially-reduced O2 species can give rise to functional and morphologic disturbances in cells. There is evidence to implicate oxygen free radicals as important pathologic mediators in many human disease processes.
Collapse
Affiliation(s)
- M Martínez-Cayuela
- Department of Biochemistry and Molecular Biology, Facultad de Farmacia, University of Granada, Spain
| |
Collapse
|
19
|
Brent JA, Rumack BH. Role of free radicals in toxic hepatic injury. II. Are free radicals the cause of toxin-induced liver injury? JOURNAL OF TOXICOLOGY. CLINICAL TOXICOLOGY 1993; 31:173-96. [PMID: 8433412 DOI: 10.3109/15563659309000384] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- J A Brent
- Rocky Mountain Poison and Drug Center, Denver, CO 80204
| | | |
Collapse
|
20
|
Vermeulen NP, Bessems JG, Van de Straat R. Molecular aspects of paracetamol-induced hepatotoxicity and its mechanism-based prevention. Drug Metab Rev 1992; 24:367-407. [PMID: 1628537 DOI: 10.3109/03602539208996298] [Citation(s) in RCA: 179] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- N P Vermeulen
- Department of Pharmacochemistry, Faculty of Chemistry, Vrije Universiteit, Amsterdam, The Netherlands
| | | | | |
Collapse
|
21
|
Nakae D, Yoshiji H, Yamamoto K, Maruyama H, Kinugasa T, Takashima Y, Denda A, Konishi Y. Influence of timing of administration of liposome-encapsulated superoxide dismutase on its prevention of acetaminophen-induced liver cell necrosis in rats. ACTA PATHOLOGICA JAPONICA 1990; 40:568-73. [PMID: 2239312 DOI: 10.1111/j.1440-1827.1990.tb01601.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The possible participation of acute oxidative stress in the in vivo mechanism by which acetaminophen (APAP) induces hepatocellular injury was examined. Male Sprague-Dawley rats were administered 3-methylcholanthrene, fasted for 18 h, then given APAP and sacrificed after a further 6 h of fasting. Extensive centrilobular liver cell necrosis along with markedly elevated serum activity of aminotransferases was observed. Liposome-encapsulated human recombinant Cu-Zn superoxide dismutase (LSOD) administered 1 or 0.5 h prior to APAP or simultaneously with the toxin completely prevented APAP-induced hepatocellular injury. In contrast, LSOD administered 5 or 2.5 h before or 1, 2.5 or 5 h after the toxin treatment did not prevent APAP toxicity. Incomplete protection against APAP-induced injury was obtained when LSOD was administered 0.5 h after the toxin. These results support the proposal of an oxidative mechanism for APAP hepatotoxicity.
Collapse
Affiliation(s)
- D Nakae
- Department of Oncological Pathology, Nara Medical University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Kaneo Y, Fujihara Y, Tanaka T, Kozawa Y, Mori H, Iguchi S. Intrahepatic delivery of glutathione by conjugation to dextran. Pharm Res 1989; 6:1025-31. [PMID: 2482968 DOI: 10.1023/a:1015922303051] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Glutathione was covalently attached to dextran (T-40) by the CNBr activation method. The compound obtained was a water-soluble powder containing 10 (w/w%) glutathione, which was gradually released from the conjugate in aqueous media. Mice depleted of glutathione by treatment with buthionine sulfoximine, a potent inhibitor of gamma-glutamylcysteine synthetase, exhibited a significant increase in hepatic glutathione level after intravenous injection of the conjugate. In mice given a lethal dose of acetaminophen, the survival rate increased progressively with coadministration of the conjugate, whereas little improvement was found when free glutathione was given. The conjugate maintained the serum transaminase activities at lower level after acetaminophen administration. These findings suggest that the dextran conjugate of glutathione is transported into hepatic cells and is intracellularly hydrolyzed to free form, which protects mice from hepatotoxicity due to acetaminophen.
Collapse
Affiliation(s)
- Y Kaneo
- Department of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Phornchirasilp S, DeSouza JJ, Feller DR. In vivo and in vitro studies of the hepatotoxic effects of 4-chlorophenol in mice. Biochem Pharmacol 1989; 38:961-72. [PMID: 2930597 DOI: 10.1016/0006-2952(89)90287-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
4-Chlorophenol (4-CP) was studied for its toxicological effect on liver by using both in vivo and in vitro approaches. Male mice were administered 4-CP, 1.5 mmol/kg body weight, i.p., and were killed at 10, 20, 30 and 50 min after drug injection. Either i.p. or oral 4-CP administration significantly lowered total liver thiol levels by 20-30% after 30 min and 3 hr respectively. This time-dependent effect of 4-CP after i.p. treatment was enhanced when mice were pretreated with hepatic microsomal enzyme inducers (phenobarbital, 40 mg/kg body weight, b.i.d., 7 days; and beta-naphthoflavone, 80 mg/kg body weight once daily, 4 days). Further, the microsomal cytochrome P-450 inhibitor, SKF 525-A, 75 mg/kg body weight injected i.p. to mice 30 min prior to 4-CP administration, blocked the reduction of liver thiol content produced by 4-CP. The results suggest that a chemically reactive intermediate of 4-CP may be formed in liver which is responsible for the observed decrease in liver thiol content. Other investigations were done to characterize the in vitro irreversible binding of [14C]4-CP. [14C]4-CP was bound irreversibly to mouse liver microsomal proteins in a concentration-dependent manner. Binding was NADPH dependent and gave a maximal binding of 12.0 nmol/mg protein/20 min and an apparent binding constant of 0.222 mM. [14C]-Binding of 4-CP was increased by 155 and 127% in liver microsomes of phenobarbital- and beta-naphthoflavone. SKF 525-A, and CO:O2 (4:1, v/v)] and selected nucleophilic compounds (glutathione, L-cysteine or L-lysine) significantly reduced [14C]4-CP binding to mouse liver microsomes. An epoxide hydrolase inhibitor, cyclohexene oxide, did not alter the extent of irreversible binding, whereas scavengers of superoxide anions or agents that are reported to reduce accumulation of active semiquinone and quinone species (L-ascorbic acid, superoxide dismutase or epinephrine) decreased the binding of [14C]4-CP to mouse liver microsomal proteins by 56, 31 and 92% respectively. The data suggest that semiquinone and quinone species of 4-CP may be the chemically reactive intermediates leading to the in vivo reduction of liver thiol levels. Since 4-CP is a minor contaminant and possible metabolite of clofibrate and chemically related hypolipidemic agents, 4-CP and its metabolites may be partly responsible for some of the hepatotoxic effects seen after long-term administration of this therapeutic class of drugs.
Collapse
|
24
|
Kolodziejczyk P, Reszka K, Lown JW. Enzymatic oxidative activation of 5-iminodaunorubicin. Spectrophotometric and electron paramagnetic resonance studies. Biochem Pharmacol 1989; 38:803-9. [PMID: 2539159 DOI: 10.1016/0006-2952(89)90234-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Horseradish peroxidase catalyzed oxidation of the antitumor agent 5-iminodaunorubicin by hydrogen peroxide was studied with both spectrophotometric and electron paramagnetic resonance methods. Kinetics of oxidation of the drug at pH 3, 6 and 8 were determined. Rapid formation of a nitrogen-centered free radical metabolite was demonstrated with electron paramagnetic resonance employing the 15N-labeled drug and by deuterium exchange techniques. This enzymatic oxidative activation of 5-iminodaunorubicin suggests an alternative mode of metabolism and mechanism of action of this less cardiotoxic anticancer agent. By contrast, the parent compound, daunorubicin, did not undergo oxidation by the horseradish peroxidase-hydrogen peroxide system.
Collapse
Affiliation(s)
- P Kolodziejczyk
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
25
|
Bisby RH, Tabassum N. Properties of the radicals formed by one-electron oxidation of acetaminophen--a pulse radiolysis study. Biochem Pharmacol 1988; 37:2731-8. [PMID: 3395354 DOI: 10.1016/0006-2952(88)90035-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The semi-iminoquinone radical of acetaminophen, which has previously been proposed as a possible hepatotoxic intermediate in the cytochrome P-450 catalysed oxidation of acetaminophen, has been generated and studied by pulse radiolysis. In the absence of other reactive solutes, the radical decays rapidly by second order kinetics with a rate constant (2k2) of (2.2 +/- 0.4) x 10(9) M-1 sec-1. In alkaline solutions the radical deprotonates with a pK of 11.1 +/- 0.1 to form a radical-anion, as confirmed by the effect of ionic strength on the rate of radical decay. The acetaminophen radical-anion reacts with resorcinol at high pH values, leading to the formation of a transient equilibrium from which the one-electron reduction potential of the semi-iminoquinone radical of acetaminophen is estimated to be +0.707 +/- 0.01 V at pH 7. This value predicts that acetaminophen should be oxidised by thiyl radicals. This was confirmed by pulse radiolysis experiments for reaction of the cysteinyl radical, for which rate constants of 7 x 10(6) M-1 sec-1 at pH 7 and 2.7 x 10(8) M-1 sec-1 at pH 11.3 were obtained. The reaction of O2 with the acetaminophen semi-iminoquinone radical could not be detected by pulse radiolysis, and alternative mechanisms for superoxide radical formation are discussed.
Collapse
Affiliation(s)
- R H Bisby
- Department of Biological Sciences, University of Salford, U.K
| | | |
Collapse
|
26
|
van de Straat R, Vromans RM, Bosman P, de Vries J, Vermeulen NP. Cytochrome P-450-mediated oxidation of substrates by electron-transfer; role of oxygen radicals and of 1- and 2-electron oxidation of paracetamol. Chem Biol Interact 1988; 64:267-80. [PMID: 3342453 DOI: 10.1016/0009-2797(88)90102-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The mechanism by which the hepatic cytochrome P-450 (Cyt. P-450) containing mixed-function oxidase system oxidizes the analgesic drug paracetamol (PAR) to a hepatotoxic metabolite was studied. Since previous studies excluded the possibility of oxygenation of PAR, three other mechanisms, namely direct 1-electron oxidation by a Cyt. P-450-ferrous-dioxygen complex under concomitant formation of H2O2 to N-acetyl-p-semiquinone imine (NAPSQI), direct 2-electron oxidation by a Cyt. P-450-ferric-oxene complex to N-acetyl-p-benzoquinone imine (NAPQI) and indirect oxidation by active oxygen species released from Cyt. P-450, were considered. Indirect oxidation by active oxygen species was not involved, as active oxygen scavengers such as superoxide dismutase, catalase and DMSO did not affect the oxidation of PAR in hepatic microsomes. No reaction products characteristic for a direct 1-electron oxidation of PAR by Cyt. P-450 were observed: neither NAPSQI radical formation was detectable by ESR, nor PAR-dimer formation, nor stimulation of the microsomal H2O2 production was found to occur. In fact, PAR inhibited the spontaneous microsomal H2O2 formation. Studies on the reactions of NAPSQI with glutathione (GSH) revealed that NAPSQI hardly conjugated with GSH to a 3-glutathionyl-paracetamol conjugate (PAR-GSH) conjugate. The reactions of the elusive reactive metabolite formed during microsomal oxidation of PAR in the presence of GSH closely resembled those of synthetic NAPQI: both PAR-GSH and oxidized glutathione (GSSG) formation occurred. Furthermore, in agreement with a 2-electron oxidation hypothesis, iodosobenzene-dependent oxidation of PAR by cyt. P-450 in the presence of GSH resulted in the formation of the PAR-GSH conjugate. It is concluded that bioactivation of PAR by the Cyt. P-450 containing mixed-function oxidase system consists of a direct 2-electron oxidation to NAPQI.
Collapse
Affiliation(s)
- R van de Straat
- Department of Pharmacochemistry, Molecular Toxicology, Free University, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
27
|
Kyle ME, Miccadei S, Nakae D, Farber JL. Superoxide dismutase and catalase protect cultured hepatocytes from the cytotoxicity of acetaminophen. Biochem Biophys Res Commun 1987; 149:889-96. [PMID: 3122747 DOI: 10.1016/0006-291x(87)90491-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Superoxide dismutase, catalase and mannitol prevent the killing of cultured hepatocytes by acetaminophen in the presence of an inhibitor of glutathione reductase, BCNU. Under these conditions, the cytotoxicity of acetaminophen depends upon its metabolism, since beta-naphthoflavone, an inhibitor of mixed function oxidation, prevents the cell killing. In hepatocytes made resistant to acetaminophen by pretreatment with the ferric iron chelator, deferoxamine, addition of ferric or ferrous iron restores the sensitivity to acetaminophen. In such a situation, both superoxide dismutase and catalase prevent the killing by acetaminophen in the presence of ferric iron. By contrast, catalase, but not superoxide dismutase, prevents the cell killing dependent upon addition of ferrous iron. These results document the participation of both superoxide anion and hydrogen peroxide in the killing of cultured hepatocytes by acetaminophen and suggest that hydroxyl radicals generated by an iron catalyzed Haber-Weiss reaction mediate the cell injury.
Collapse
Affiliation(s)
- M E Kyle
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107
| | | | | | | |
Collapse
|
28
|
Castell JV, Gomez MJ, Mirabet V, Miranda MA, Morera IM. Photolytic degradation of benorylate: effects of the photoproducts on cultured hepatocytes. J Pharm Sci 1987; 76:374-8. [PMID: 2888867 DOI: 10.1002/jps.2600760507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The photodegradation of benorylate [4'-(acetamido)phenyl-2-acetoxybenzoate], a drug frequently used in rheumatoid arthritis therapy, has been examined under different sets of experimental conditions. Several photoproducts have been isolated and identified on the basis of their IR, NMR, and MS spectra. The most significant photochemical process is the photo-Fries rearrangement of benorylate, leading to 5-acetamido-2'-acetoxy-2-hydroxybenzophenone (1). This compound undergoes a rapid transacylation to the isomeric 5'-acetamido-2'-acetoxy-2-hydroxybenzophenone (2). A primary culture of rat hepatocytes has been used to evaluate the possible toxicity of these two benzophenones, keeping in mind the following criteria: leakage of cytosolic enzymes, attachment index to culture plates, gluconeogenesis from lactate and fructose, glycogen balance, and albumin synthesis. At the concentrations assayed, neither of the two major photoproducts of benorylate (benzophenones 1 and 2) had significant toxic effects on liver cells in culture.
Collapse
Affiliation(s)
- J V Castell
- Centro de Investigación, Hospital La Fe, Valencia, Spain
| | | | | | | | | |
Collapse
|
29
|
van de Straat R, de Vries J, Vermeulen NP. Role of hepatic microsomal and purified cytochrome P-450 in one-electron reduction of two quinone imines and concomitant reduction of molecular oxygen. Biochem Pharmacol 1987; 36:613-9. [PMID: 3030331 DOI: 10.1016/0006-2952(87)90710-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The possible role of cytochrome P-450 in one-electron reduction of quinoid compounds as well as in the formation of reduced oxygen species was investigated in hepatic microsomal and reconstituted systems of purified cytochrome P-450 and purified NADPH-cytochrome P-450 reductase using electron spin resonance (ESR) methods. Two compounds were selected as model compounds: N-acetyl-parabenzoquinone imine (NAPQI) and 3,5-dimethyl-N-acetyl-para-benzoquinone imine (3,5-dimethyl-NAPQI). Both compounds could be reduced by oxyhaemoglobin, the semiquinones formed were detectable by ESR and did not reduce molecular oxygen. Both NAPQI and 3,5-dimethyl-NAPQI underwent one-electron reduction in microsomal systems and in fully reconstituted systems of cytochrome P-450 and NADPH-cytochrome P-450 reductase under anaerobic and aerobic conditions. In both incubation systems the semiquinone formation was diminished under aerobic circumstances and concomitant reduction of oxygen occurred, leading to the formation of hydrogen peroxide and hydroxyl free radicals. Both the reduction of the quinone imines and the reduction of oxygen were found to be cytochrome P-450 dependent. Both activities of cytochrome P-450 may also be involved in the bioactivation of other compounds with quinoid structural elements, like many chemotherapeutic agents.
Collapse
|
30
|
Abstract
Aerobic organisms by definition require oxygen, and the importance of iron in aerobic respiration has long been recognized, but despite their beneficial roles, these elements can pose a real threat to the organism. During oxygen reduction, reactive species such as O2-. and H2O2 are formed readily. Iron can combine with these species, or with molecular oxygen itself, to generate free radicals which will attack the polyunsaturated fatty acids of membrane lipids. This oxidative deterioration of membrane lipids is known as lipid peroxidation. To protect itself against this form of attack, the organism possesses several types of defense mechanisms. Under normal conditions, these defenses appear to offer adequate protection for cell membranes, but the possibility exists that certain foreign compounds may interfere with or even overwhelm these defenses, and herein could lie a general mechanism of toxicity. This possible cause of toxicity is discussed in relation to other suggested causes.
Collapse
Affiliation(s)
- A A Horton
- Department of Biochemistry, University of Birmingham, England
| | | |
Collapse
|
31
|
Potter DW, Hinson JA. Mechanisms of acetaminophen oxidation to N-acetyl-P-benzoquinone imine by horseradish peroxidase and cytochrome P-450. J Biol Chem 1987. [DOI: 10.1016/s0021-9258(19)75735-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
32
|
Van De Straat R, De Vries J, Kulkens T, Debets AJ, Vermeulen NP. Paracetamol, 3-monoalkyl- and 3,5-dialkyl derivatives. Comparison of their microsomal cytochrome P-450 dependent oxidation and toxicity in freshly isolated hepatocytes. Biochem Pharmacol 1986; 35:3693-9. [PMID: 3778500 DOI: 10.1016/0006-2952(86)90653-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The effects of 3-monoalkyl- and 3,5-dialkyl-substitution on the cytotoxicity of paracetamol (PAR) in rat hepatocytes was studied. PAR is known to be bioactivated by the hepatic microsomal cytochrome P-450 containing a mixed-function oxidase system presumably to N-acetyl-para-benzoquinone imine (NAPQI), a reactive metabolite which upon overdosage of the drug causes depletion of cellular glutathione (GSH) and hepatotoxicity. The four 3-mono- and the four 3,5-di-alkyl-substituted derivatives of PAR investigated in this study (R = CH3, C2H5, C3H7, C4H9) interacted with cytochrome P-450 giving rise to reverse type I spectral changes. Like PAR, all derivatives underwent cytochrome P-450-mediated oxidation to NAPQIs. In contrast to induction by phenobarbital, induction of cytochrome P-450 by 3-methylcholanthrene enhanced the microsomal oxidation of PAR and its derivatives. The NAPQIs formed from PAR and the 3-mono-alkyl derivatives by microsomal oxidation were found to conjugate with GSH and to oxidise GSH to GSSG. The NAPQIs formed from the 3,5-dialkyl-substituted derivatives, however, only oxidized GSH to GSSG. PAR and the 3-monoalkyl derivatives were found to deplete cellular GSH to about the same extent and to be equally toxic in freshly isolated hepatocytes from 3-methylcholanthrene treated rats. In contrast, the 3,5-di-alkyl-substituted derivatives of PAR did not affect the GSH levels and were not toxic in the hepatocytes, even at higher concentrations. It is suggested that the difference between the way of reacting of 3,5-dialkyl-NAPQIs and NAPQIs from PAR and 3-monoalkyl derivatives with thiols of cellular GSH and protein could account for the observed difference between the toxicity of the 3,5-dialkyl- and the 3-monoalkyl-substituted derivatives of PAR.
Collapse
|
33
|
Wendel A, Hallbach J. Quantitative assessment of the binding of acetaminophen metabolites to mouse liver microsomal phospholipid. Biochem Pharmacol 1986; 35:385-9. [PMID: 3947378 DOI: 10.1016/0006-2952(86)90209-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Phospholipids were quantitatively extracted from microsomes and separated by an h.p.l.c. gradient system with a solvent mixture of n-hexane/n-propanol/water/acetic acid. In a model reaction using horseradish peroxidase/H2O2 in order to activate acetaminophen and inactivated microsomes as target, a covalent binding of 10 nmol drug metabolite per mg microsomal lipid was found. In isolated intact microsomes from methylcholanthrene-pretreated male albino mice, a binding of 0.1 nmol acetaminophen metabolite per mg phospholipid was determined while the binding of metabolites to protein amounted to 3 nmol/mg. The results demonstrate that in mouse liver microsomes metabolizing acetaminophen, about one out of 10(4) phospholipid molecules is modified.
Collapse
|
34
|
Potter DW, Hinson JA. Reactions of glutathione with oxidative intermediates of acetaminophen. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1986; 197:763-72. [PMID: 3766293 DOI: 10.1007/978-1-4684-5134-4_71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
|
36
|
Abstract
A variety of enzymes function in the oxygenation, oxidation-reduction, conjugation, and hydrolysis of drugs and other foreign chemicals. Often these enzymes detoxicate chemicals to prevent detrimental effects. In this review we will, however, concentrate on cases in which metabolism activates chemicals to reactive species which cause cellular damage. Particular attention will be given to mixed-function oxidases, which carry out a variety of oxygenations, as well as other reactions. (We will focus on cellular toxicity as opposed to initiation of tumorigenesis in this review.) In many cases, considerable circumstantial evidence exists linking these enzymes to enhanced toxicity of chemicals, although causal relationships have seldom been demonstrated. Further, in very few cases is the explicit cause of toxicity known. Modification of critical protein residues is suspected, although oxidative stress may also be involved in some cases. We discuss general aspects of mechanisms of toxic action, briefly list all cases in which metabolism is suspected to play a role in enhancing toxicity, and review a few examples in detail where substantial chemical and enzymatic information is available. The latter instances would involve knowledge of the enzymes involved, chemical evidence on the structures of the reactive metabolites, identification of adducts, and some inference into the biological processes which are effected to elicit toxicity. We consider, in this regard, vinyl halides (which have been a focus in our own laboratory), acetaminophen, pyrrolizidine alkaloids, and fluoroxene.
Collapse
|
37
|
Lunte SM, Kissinger PT. The use of liquid chromatography with dual-electrode electrochemical detection in the investigation of glutathione oxidation during benzene metabolism. J Chromatogr A 1984; 317:579-88. [PMID: 6530454 DOI: 10.1016/s0021-9673(01)91698-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Liquid chromatography with electrochemical detection provides a powerful tool for the study of glutathione oxidation during benzene metabolism. The use of two dual-electrode detectors allows for the detection of oxidized and reduced glutathione and of phenol and quinone metabolites of benzene. The role of glutathione as a reductant is explored in this paper. Results indicate that hydrogen peroxide is the oxidizing agent primarily responsible for glutathione oxidation during benzene, phenol and hydroquinone metabolism.
Collapse
|
38
|
West PR, Harman LS, Josephy PD, Mason RP. Acetaminophen: enzymatic formation of a transient phenoxyl free radical. Biochem Pharmacol 1984; 33:2933-6. [PMID: 6089841 DOI: 10.1016/0006-2952(84)90222-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
39
|
Fischer V, Mason RP. Stable free radical and benzoquinone imine metabolites of an acetaminophen analogue. J Biol Chem 1984. [DOI: 10.1016/s0021-9258(18)90961-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
40
|
Savides MC, Oehme FW, Nash SL, Leipold HW. The toxicity and biotransformation of single doses of acetaminophen in dogs and cats. Toxicol Appl Pharmacol 1984; 74:26-34. [PMID: 6729821 DOI: 10.1016/0041-008x(84)90266-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The biotransformation of single oral doses of acetaminophen (APAP) was studied in dogs an cats. Each animal received APAP at a no-effect (low), mildly toxic (medium), and severely toxic (high) dosage; dosages for each species were selected to produce similar clinical effects at each respective dosage. For dogs, these dosages were 100, 200, and 500 mg APAP/kg, while for cats, the similar effective dosages were 20, 60, and 120 mg APAP/kg. Plasma half-lives in dogs remained constant at the lower two dosages, but nearly tripled at the high dosage. The plasma half-lives in cats rose with increased dosage. Although the cats were given lower APAP dosages than the dogs, the plasma half-lives of cats were greater than those of the dogs at the medium and high dosages. Both species excreted about 85% of the administered single dose within the first 24 hr. APAP-glucuronide was the principal metabolite excreted in the urine of dogs; its fraction of the total metabolites excreted in urine remained constant at the three dose levels. In cats, APAP-sulfate was the major metabolite in urine at all three dosage levels, but the fraction of the total urinary metabolites represented by APAP-sulfate decreased as the dosage increased. Hepatic centrilobular pathology was seen in dogs, while cats had more diffuse liver pathologic changes. The results indicate that the cat is at increased risk from APAP exposure because of impaired glucuronidation and saturation of its sulfate conjugation pathway.
Collapse
|
41
|
De Vries J, De Jong J, Lock FM, Van Bree L, Mullink H, Veldhuizen RW. Protection against paracetamol-induced hepatotoxicity by acetylsalicylic acid in rats. Toxicology 1984; 30:297-304. [PMID: 6729828 DOI: 10.1016/0300-483x(84)90140-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Acetylsalicylic acid (ASA) given simultaneously with paracetamol decreased paracetamol-induced hepatotoxicity (measured by plasma transaminase activities as well as histology) without any effect on glutathione depletion, indicating that ASA prevents a process (or processes) subsequent to the metabolic activation of paracetamol. Delayed treatment with ASA also reduced paracetamol-induced liver toxicity, suggesting that reduction of the absorption rate of paracetamol does not contribute essentially to the protection by ASA. Combinations of paracetamol and ASA may have potential use in the development of safer analgesic combinations containing paracetamol (or ASA).
Collapse
|
42
|
Eyer P, Lengfelder E. Radical formation during autoxidation of 4-dimethylaminophenol and some properties of the reaction products. Biochem Pharmacol 1984; 33:1005-13. [PMID: 6324808 DOI: 10.1016/0006-2952(84)90507-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
4-Dimethylaminophenol (DMAP), after intravenous injection, rapidly forms ferrihaemoglobin and has been successfully used in the treatment of cyanide poisoning. Since DMAP produces many equivalents of ferrihaemoglobin, it was of interest to obtain further insight into this catalytic process. DMAP autoxidizes readily at pH regions above neutrality, a process which is markedly accelerated by oxyhaemoglobin. The resulting red-coloured product was identified as the 4-(N,N-dimethylamino) phenoxyl radical by EPR spectroscopy. The same radical was also produced by pulse radiolysis and oxidation with ferricyanide. The 4-(N,N-dimethylamino)phenoxyl radical is quite unstable and decays in a pseudo-first order reaction (k = 0.4 sec-1 at pH 8.5, 22 degrees) with the formation of p-benzoquinone and dimethylamine. This observed decay rate is identical with the rate of hydrolysis of N,N-dimethylquinonimine. When a solution containing the phenoxyl radical was extracted with ether, half the stoichiometric amount of DMAP was recovered. Hence it is apparent that the phenoxyl radical decays by disproportionation yielding DMAP and N,N-dimethylquinonimine. The latter product then quickly hydrolyses. The equilibrium of this disproportionation reaction is far towards the radical side, and the pseudo-first order hydrolysis controls the radical decay rate. p-Benzoquinone rapidly reacts with DMAP (k2 = 2 X 10(4) M-1 sec-1) with the formation of the 4-(N,N-dimethylamino)phenoxyl and the semiquinone radicals. This reaction explains the autocatalytic phenoxyl radical formation during autoxidation of DMAP. DMAP is not oxidized by H2O2 or O-.2 but the 4-(N,N-dimethylamino)phenoxyl radical is very rapidly reduced by O-.2 (k2 = 2 X 10(8) M-1 sec-1). In addition, the phenoxyl radical is quickly reduced by NAD(P)H or GSH with the formation of NAD(P)+ or GSSG. Since DMAP is also able to reduce two equivalents of ferrihaemoglobin (provided that the ferrohaemoglobin produced is trapped by carbon monoxide), electrophilic addition reactions of the phenoxyl radical seem unimportant in contrast to N,N-dimethylquinonimine. Hence, during the catalytic ferrihaemoglobin formation, DMAP is oxidized by oxygen which is activated by haemoglobin, and the phenoxyl radical oxidizes ferrohaemoglobin. This catalytic process is terminated by covalent binding of N,N-dimethylquinonimine to SH groups of haemoglobin (and GSH in red cells).
Collapse
|
43
|
Nakagawa Y, Hiraga K, Suga T. On the mechanism of covalent binding of butylated hydroxytoluene to microsomal protein. Biochem Pharmacol 1983; 32:1417-21. [PMID: 6860361 DOI: 10.1016/0006-2952(83)90456-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The structures of cysteine conjugates of 3,5-di-tert-butyl-4-hydroxytoluene (BHT) and the binding sites of BHT metabolites on microsomal protein were investigated by 13C nuclear magnetic resonance (13C-NMR) and gas-liquid chromatography/mass spectrometry. The cysteine conjugates of 2,6-di-tert-butyl-4-hydroxymethylphenol (BHT-alcohol) and 2,6-di-tert-butyl-4-methylene-2,5-cyclohexadienone (quinone methide), which are metabolites of BHT found in rat liver and specifically reacts with thiol compounds, were prepared as alcoholic aqueous solutions. The molecular structure of the cysteine conjugate of BHT-alcohol agreed completely with that of quinone methide in 13C-NMR spectra or mass spectra. These spectra of both conjugates further showed that the conjugates are due to thioether binding between the 4-methyl group of metabolites and the sulfhydryl group of cysteine. When [14C]BHT-bound microsomes prepared in vitro were enzymatically hydrolyzed with Pronase E, the major radioactive material that eluted with methanol from a column of Amberlite XAD-2 and gave a positive ninhydrin reaction was identified as a cysteine conjugate of BHT by comparing its Rf values on TLC and mass spectrum. On the basis of the results, it was apparent that the binding site of activated substituents of BHT on protein was mainly the sulfhydryl group of cysteine residue.
Collapse
|
44
|
Abstract
Acetaminophen (APAP) is considered one of the safest of all minor analgesics, but when taken in large doses (greater than 10 g) toxicity occurs. Severely poisoned patients experience hepatic and/or renal failure. The major metabolic pathway of APAP is formation of glucuronide and sulfate conjugates. A minor pathway is formation of a reactive metabolite that conjugates with glutathione (GSH). When GSH is depleted, the reactive metabolite causes necrosis of hepatic and other tissues. Treatment of APAP toxicity involves supplying alternate sulfhydryl donors or inhibiting oxidative formation of the reactive metabolite. Estimation of plasma APAP levels is necessary for effective treatment.
Collapse
|
45
|
Reiter R, Wendel A. Drug-induced lipid peroxidation in mice--IV. In vitro hydrocarbon evolution, reduction of oxygen and covalent binding of acetaminophen. Biochem Pharmacol 1983; 32:665-70. [PMID: 6830629 DOI: 10.1016/0006-2952(83)90491-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
46
|
|
47
|
Abstract
Most cases of chemical toxicity involve one or both of the fundamental pathological processes of "acute lethal injury" and "autoxidative cellular injury." The process of "acute lethal injury" by which toxic chemicals interfere with cellular energy metabolism, leading ultimately to cell death and tissue necrosis, is well known and reasonably understood. Inhibition of glycolysis, mitochondrial respiration, or oxidative phosphorylation, resulting in lack of ATP synthesis or inhibition of ATPase and other enzymes, leads to decreased efficiency of the sodium pump, hydropic degeneration, lipid accumulation, and eventually cell death. Less well known are the mechanisms whereby toxic chemicals initiate autoxidation, leading to "autoxidative cellular injury," disrupting cell membranes, and resulting in increased autophagocytosis, cell death, and mutations. Many reactive intermediates of toxic chemicals are electrophiles, free radicals, or free-radical generators, which may potentiate the toxicity of tissue oxygen, depleting intracellular glutathione and biological antioxidants, resulting in membrane damage, impairment of the calcium pump, cell death, and damage to DNA. The mechanisms of oxygen toxicity and chemical-mediated oxygen toxicity are discussed, with particular reference to the microsomal mixed-function oxidase system and its role in the detoxication and activation of environmental chemicals. The dependence of tissue oxygen concentration, the rates of oxidative activation of chemicals, and the extents of autoxidative cellular injury on the size of the animal species is considered, and the importance of this to the scientific evaluation of chemical toxicity is discussed.
Collapse
|
48
|
Moldéus P, Andersson B, Rahimtula A, Berggren M. Prostaglandin synthetase catalyzed activation of paracetamol. Biochem Pharmacol 1982; 31:1363-8. [PMID: 6807320 DOI: 10.1016/0006-2952(82)90029-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Prostaglandin synthetase has the ability to catalyze the metabolism of paracetamol to a reactive metabolite, which binds to protein and reduced glutathione (GSH). This was demonstrated with microsomes isolated from both sheep seminal vesicles (SSV) and rabbit kidney medulla. The activation of paracetamol occurred through cooxygenation during prostaglandin biosynthesis, the peroxidase activity of this enzyme being responsible for the reaction. In addition to being metabolized, paracetamol also stimulated the rate of prostaglandin biosynthesis probably by serving as a potent hydrogen donor. The metabolism of paracetamol to a reactive metabolite most likely involved the formation of a paracetamol radical species. This was indicated by an inhibitory effect of the antioxidant, butylated hydroxyanisole, as well as by a very rapid oxidation of GSH during the course of the prostaglandin synthetase catalyzed reaction. Whether this paracetamol radical is further oxidized to the quinone imine prior to reacting with GSH or protein, remains to be established. The ultimate reactive metabolite is evidently the same as that formed with liver microsomes and NADPH since the glutathione conjugates were apparently identical. The rate of paracetamol activation by SSV microsomes was, however, more than 100 times that by liver microsomes and furthermore the apparent Km was considerably lower. Finally, N-OH paracetamol was shown to be activated by prostaglandin synthetase in the presence of arachidonic acid to a metabolite apparently different from that formed from paracetamol.
Collapse
|
49
|
Chapter 33. Drug Metabolism. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 1982. [DOI: 10.1016/s0065-7743(08)60515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
50
|
|