1
|
Cui Q, Jiang T, Xie X, Wang H, Qian L, Cheng Y, Li Q, Lu T, Yao Q, Liu J, Lai B, Chen C, Xiao L, Wang N. S-nitrosylation attenuates pregnane X receptor hyperactivity and acetaminophen-induced liver injury. JCI Insight 2024; 9:e172632. [PMID: 38032737 PMCID: PMC10906221 DOI: 10.1172/jci.insight.172632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Drug-induced liver injury (DILI), especially acetaminophen overdose, is the leading cause of acute liver failure. Pregnane X receptor (PXR) is a nuclear receptor and the master regulator of drug metabolism. Aberrant activation of PXR plays a pathogenic role in the acetaminophen hepatotoxicity. Here, we aimed to examine the S-nitrosylation of PXR (SNO-PXR) in response to acetaminophen. We found that PXR was S-nitrosylated in hepatocytes and the mouse livers after exposure to acetaminophen or S-nitrosoglutathione (GSNO). Mass spectrometry and site-directed mutagenesis identified the cysteine 307 as the primary residue for S-nitrosylation (SNO) modification. In hepatocytes, SNO suppressed both agonist-induced (rifampicin and SR12813) and constitutively active PXR (VP-PXR, a human PXR fused to the minimal transactivator domain of the herpes virus transcription factor VP16) activations. Furthermore, in acetaminophen-overdosed mouse livers, PXR protein was decreased at the centrilobular regions overlapping with increased SNO. In PXR-/- mice, replenishing the livers with the SNO-deficient PXR significantly aggravated hepatic necrosis, increased HMGB1 release, and exacerbated liver injury and inflammation. Particularly, we demonstrated that S-nitrosoglutathione reductase (GSNOR) inhibitor N6022 promoted hepatoprotection by increasing the levels of SNO-PXR. In conclusion, PXR is posttranslationally modified by SNO in hepatocytes in response to acetaminophen. This modification mitigated the acetaminophen-induced PXR hyperactivity. It may serve as a target for therapeutical intervention.
Collapse
Affiliation(s)
- Qi Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Tingting Jiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Xinya Xie
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Haodong Wang
- East China Normal University Health Science Center, Shanghai, China
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yanyan Cheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Qiang Li
- School of Public Health, Xi’an Jiaotong University, Xi’an, China
| | - Tingxu Lu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Qinyu Yao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Jia Liu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Baochang Lai
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lei Xiao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Nanping Wang
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
2
|
Rofaeil RR, Welson NN, Fawzy MA, Ahmed AF, Atta M, Bahaa El-Deen MA, Abdelzaher WY. The IL-6/HO-1/STAT3 signaling pathway is implicated in the amelioration of acetaminophen-induced hepatic toxicity: A neonatal rat model. Hum Exp Toxicol 2023; 42:9603271231151376. [PMID: 36625353 DOI: 10.1177/09603271231151376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The widespread use of acetaminophen (APAP) in children as an over-the-counter treatment can cause acute liver failure through accidental overdose or ingestion. Therefore, the current research sought to investigate the function of hemin in mitigating the acute hepatotoxic effect of APAP in rat offspring. Thirty-two rats were assigned into four groups: control, hemin, APAP, and hemin/APAP groups. Liver enzymes were measured in serum along with oxidative stress indicators, tumor necrosis factor-α (TNF-α), interleukin-1beta (IL-1β), total nitrites (NOx), and caspase 3 in liver. Immunoblotting of heme oxygenase-1 (HO-1), interleukin-6 (IL-6), Janus kinase 2 (Jak2), and signal transducer and activator of transcription 3 (STAT3) was carried out. The Bax/Bcl2 mRNA expression ratio was determined. A histological study and an immunohistochemical study of phosphorylated STAT3 were also done. Hemin reduced liver enzymes, MDA, TNF-α, NOx, caspase 3, IL-1β, p-STAT3 expression, p-Jak2 expression, IL-6 expression, and Bax/Bcl2 mRNA expression ratio. In contrast, hemin increased GSH, TAC, and the expression of HO-1, improving the histopathological picture of liver tissue. Thus, hemin could ameliorate APAP-induced hepatic toxicity in rat offspring through anti-oxidant, anti-apoptotic, and anti-inflammatory actions with a possible role for the IL-6/HO-1/Jak2/STAT3 pathway.
Collapse
Affiliation(s)
- Remon Roshdy Rofaeil
- Department of Pharmacology, Faculty of Medicine, 68877Minia University, Minia, Egypt.,Department of Pharmacology, Deraya University, New Minia City, Egypt
| | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, 158411Beni-Suef University, Beni-Suef, Egypt
| | - Michael A Fawzy
- Department of Biochemistry, Faculty of Pharmacy, 68877Minia University, Minia, Egypt
| | - Amira F Ahmed
- Department of Histology and Cell Biology, Faculty of Medicine, 68877Minia University, Minia, Egypt.,Department of Histology and Cell Biology, Misr University for Science and Technology, October City, Egypt
| | - Medhat Atta
- Department of Anatomy, Faculty of Medicine, 68877Minia University, Minia, Egypt
| | | | | |
Collapse
|
3
|
Vanova J, Malinak D, Andrys R, Kubat M, Mikysek T, Rousarova E, Musilek K, Rousar T, Cesla P. Optimization of Gradient Reversed Phase High Performance Liquid Chromatography Analysis of Acetaminophen Oxidation Metabolites using Linear and Non-linear Retention Model. J Chromatogr A 2022; 1669:462956. [DOI: 10.1016/j.chroma.2022.462956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 11/15/2022]
|
4
|
Pietruk K, Gbylik-Sikorska M, Łebkowska-Wieruszewska B, Gajda A, Giorgi M, Sartini I, Jedziniak P. Development of a Multimatrix UHPLC-MS/MS Method for the Determination of Paracetamol and Its Metabolites in Animal Tissues. Molecules 2021; 26:molecules26072046. [PMID: 33918518 PMCID: PMC8038326 DOI: 10.3390/molecules26072046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Paracetamol/acetaminophen (APAP) is one of the most popular pharmacologically active substances used as an analgesic and antipyretic agent. The metabolism of this drug occurs in the liver and leads to the formation of two main metabolites-glucuronic acid and sulfate derivate. Despite the wide use of paracetamol in veterinary medicine, a handful of analytical methods were published for the determination of paracetamol residues in animal tissues. In this paper, a multimatrix method has been developed for the determination of paracetamol and two metabolites-paracetamol sulfate (PS) and p-Acetamidophenyl β-D-glucuronide (PG). A validation procedure was conducted to verify method reliability and fit purpose as a tool for analyzing acetaminophen and metabolites in muscle, liver, lung, and kidney samples from different species of animals. Established validation parameters were in agreement with acceptable criteria laid by the European legislation. The initial significant matrix effect was successfully reduced by implementing an internal standard-4-Acetamidophenyl β-D-glucuronide-d3 (PG-d3, IS). The usefulness of the developed method was verified by analyzing samples from an experiment in which paracetamol was administrated to geese.
Collapse
Affiliation(s)
- Konrad Pietruk
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, 24-100 Pulawy, Poland; (M.G.-S.); (A.G.); (P.J.)
- Correspondence: ; Tel.: +48 81-889-3169
| | - Małgorzata Gbylik-Sikorska
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, 24-100 Pulawy, Poland; (M.G.-S.); (A.G.); (P.J.)
| | - Beata Łebkowska-Wieruszewska
- Department of Pharmacology, Toxicology and Environmental Protection, University of Life Sciences, 20-950 Lublin, Poland;
| | - Anna Gajda
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, 24-100 Pulawy, Poland; (M.G.-S.); (A.G.); (P.J.)
| | - Mario Giorgi
- Department of Veterinary Sciences, University of Pisa, 56126 Pisa, Italy;
- Department of Veterinary Medicine, PhD School, University of Sassari, 07100 Sassari, Italy;
| | - Irene Sartini
- Department of Veterinary Medicine, PhD School, University of Sassari, 07100 Sassari, Italy;
| | - Piotr Jedziniak
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, 24-100 Pulawy, Poland; (M.G.-S.); (A.G.); (P.J.)
| |
Collapse
|
5
|
AKBAŞ İ, KOCAK AO, GÜÇLÜ UTLU S, DOĞRUYOL S, ÇAKIR Z. Paracetamol Overdose May Cause Transudative Pleural Effusion in Adults. JOURNAL OF EMERGENCY MEDICINE CASE REPORTS 2021. [DOI: 10.33706/jemcr.834306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Yang JH, Na CS, Cho SS, Kim KM, Lee JH, Chen XQ, Ku SK, Cho IJ, Kim EJ, Lee JH, Ki SH. Hepatoprotective Effect of Neoagarooligosaccharide via Activation of Nrf2 and Enhanced Antioxidant Efficacy. Biol Pharm Bull 2020; 43:619-628. [PMID: 32009027 DOI: 10.1248/bpb.b19-00697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neoagarooligosaccharides (NAOS) are generated by β-agarases, which cleave the β-1,4 linkage in agarose. Previously, we reported that NAOS inhibited fat accumulation in the liver and decreased serum cholesterol levels. However, the hepatoprotective effect of NAOS on acute liver injury has not yet been investigated. Thus, we examined whether NAOS could activate nuclear factor (NF)-E2-related factor 2 (Nrf2)-antioxidant response element (ARE) and upregulates its target gene, and has hepatoprotective effect in vivo. In hepatocytes, phosphorylation and subsequent nuclear translocation of Nrf2 are increased by treatment with NAOS, in a manner dependent on p38 and c-Jun N-terminal kinase (JNK). Consistently, NAOS augmented ARE reporter gene activity and the antioxidant protein levels, resulting in increased intracellular glutathione levels. NAOS antagonized tert-butylhydroperoxide-induced reactive oxygen species (ROS) generation. Moreover, NAOS inhibited acetaminophen (APAP)-induced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and significantly decreased hepatocyte degeneration and inflammatory cell infiltration. Moreover, ROS production and glutathione depletion by APAP were reversed by NAOS. APAP-mediated apoptotic signaling pathways were also inhibited in NAOS-treated mice. Upregulalted hepatic expression of genes related to inflammation by APAP were consistently diminished by NAOS. Collectively, our results demonstrate that NAOS exhibited a hepatoprotective effect against APAP-mediated acute liver damage through its antioxidant capacity.
Collapse
Affiliation(s)
- Ji Hye Yang
- College of Pharmacy, Chosun University.,College of Korean Medicine, Dongshin University
| | - Chang-Su Na
- College of Korean Medicine, Dongshin University
| | | | | | | | - Xi-Qiang Chen
- College of Pharmacy, Chosun University.,Lab of Drug Screening, Biology Institute of Shandong Academy of Sciences
| | - Sae Kwang Ku
- MRC-GHF, College of Korean Medicine, Daegu Haany University
| | - Il Je Cho
- MRC-GHF, College of Korean Medicine, Daegu Haany University
| | | | | | | |
Collapse
|
7
|
Li FC, Lee SL, Lin HM, Lin CJ, Wang SS, Lee YY, Lo SY, Sun TL, Chen WL, Lo W, Horton N, Xu C, Chiang SJ, Chen YF, Lee HS, Dong CY. Dynamic visualization of the recovery of mouse hepatobiliary metabolism to acetaminophen-overdose damage. JOURNAL OF BIOPHOTONICS 2019; 12:e201800296. [PMID: 30302934 DOI: 10.1002/jbio.201800296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 06/08/2023]
Abstract
Acetaminophen (APAP) overdose is one of the world's leading causes of drug-induced hepatotoxicity. Although traditional methods such as histological imaging and biochemical assays have been successfully applied to evaluate the extent of APAP-induced liver damage, detailed effect of how APAP overdose affect the recovery of hepatobiliary metabolism and is not completely understood. In this work, we used intravital multiphoton microscopy to image and quantify hepatobiliary metabolism of the probe 6-carboxyfluorescein diacetate in APAP-overdose mice. We analyzed hepatobiliary metabolism for up to 7 days following the overdose and found that the excretion of the probe molecule was the most rapid on Day 1 following APAP overdose and slowed down on Days 2 and 3. On Day 7, probe excretion capability has exceeded that of the normal mice, suggesting that newly regenerated hepatocytes have higher metabolic capabilities. Our approach may be further developed applied to studying drug-induced hepatotoxicity in vivo.
Collapse
Affiliation(s)
- Feng-Chieh Li
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Sheng-Lin Lee
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Hung-Ming Lin
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Chih-Ju Lin
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Sheng-Shun Wang
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yu-Yang Lee
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Su-Yen Lo
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Tzu-Lin Sun
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Wen Lo
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Nicholas Horton
- School of Engineering and Applied Physics, Cornell University, Ithaca, New York
| | - Chris Xu
- School of Engineering and Applied Physics, Cornell University, Ithaca, New York
| | - Shu-Jen Chiang
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yang-Fang Chen
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Hsuan-Shu Lee
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan, Republic of China
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Chen-Yuan Dong
- Department of Physics, National Taiwan University, Taipei, Taiwan, Republic of China
- Center for Optoelectronic Biomedicine, National Taiwan University, Taipei, Taiwan, Republic of China
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan, Republic of China
| |
Collapse
|
8
|
Li N, Kong M, Zeng S, Xu Z, Li M, Hong W, Chu X, Sun X, Zhu M, Xu Y. The chromatin remodeling protein BRG1 regulates APAP-induced liver injury by modulating CYP3A11 transcription in hepatocyte. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3487-3495. [PMID: 30293568 DOI: 10.1016/j.bbadis.2018.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/12/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
Acetaminophen (APAP) overdose represents the most frequent cause of acute liver failure. The underlying epigenetic mechanism is not fully understood. In the present study we investigated the mechanism whereby the chromatin remodeling protein brahma related gene 1 (Brg1) regulates APAP induced liver injury in mice. We report that hepatocyte-specific deletion of Brg1 attenuated APAP induced liver injury in mice as evidenced by reduced plasma ALT and AST levels, decreased liver necrosis, amelioration of GSH depletion, and prolonged survival. Brg1 regulated APAP-induced liver injury likely by stimulating the transcription of Cyp3a11, a key cytochrome enzyme involved in APAP metabolism. Immunoprecipitation coupled with DNA affinity microarray identified hepatocyte nuclear factor 4 (HNF4) as a novel binding partner for Brg1. HNF4 recruited Brg1 to the Cyp3a11 promoter and formed a complex with Brg1 to trans-activate Cyp3a11. In contrast, BRG1 deficiency attenuated HNF4 binding to the Cyp3a11 promoter and dampened Cyp3a11 transcription. Therefore, our data suggest that Brg1 might play an essential role mediating APAP induced liver injury in vivo.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Sheng Zeng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Min Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenxuan Hong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xuehui Chu
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xitai Sun
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Min Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Department of Anatomy, Nanjing Medical University, Nanjing, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research, Liaocheng University, Liaocheng, China.
| |
Collapse
|
9
|
Politakis N, Belavgeni A, Efthimiou I, Charalampous N, Kourkouta C, Dailianis S. The impact of expired commercial drugs on non-target marine species: A case study with the use of a battery of biomarkers in hemocytes of mussels. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 148:160-168. [PMID: 29045922 DOI: 10.1016/j.ecoenv.2017.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/18/2017] [Accepted: 10/06/2017] [Indexed: 06/07/2023]
Abstract
The present study investigated the effects of two expired commercial medicines, like Buscopan Plus and Mesulid, commonly classified as household medical wastes, on hemocytes of mussel Mytilus galloprovincialis. Mussel hemocytes' lysosomal membrane stability (in terms of neutral red retention assay), superoxide anions (O2·-) and nitric oxides (NO, in terms of nitrites) production, lipid peroxidation (in terms of malondialdehyde/MDA content) and the formation of nuclear abnormalities (using the micronucleus/MN assay) were assessed in hemocytes of mussels treated for 7 days with appropriate amounts of each drug (the concentrations of active substances were considered in each case, due to the absence of data related with the excipients) as well as in hemocytes of post-treated/recovered mussels (7 days post-treatment/recovery period). According to the results, treated mussels showed significantly decreased NRRT values, enhanced O2·-, NO and MDA levels, as well as high frequencies of nuclear abnormalities in both cases. Thοse effects showed a drastic reduction in almost all cases, after the post-treatment/recovery period. Moreover, the "stress on stress" method, commonly performed for estimating mussels' ability to survive in air, showed significantly reduced LT50 values in challenged mussels, compared to values observed in control mussels. The current findings revealed for the first time that both expired commercial drugs could affect mussels, probably via the formation of active substances bioactivated metabolites, as well as excipients, such as TiO2 and SiO2, at least in case of Buscopan plus. Although further research is needed, the current findings indicate the environmental impact of expired commercial drugs, thus revealing the need for the proper disposal of household medical wastes.
Collapse
Affiliation(s)
- Nektarios Politakis
- Section of Animal Biology, Department of Biology, Faculty of Sciences, University of Patras, Rio, GR-26500 Patra, Greece
| | - Alexia Belavgeni
- Section of Animal Biology, Department of Biology, Faculty of Sciences, University of Patras, Rio, GR-26500 Patra, Greece
| | - Ioanna Efthimiou
- Department of Environmental and Natural Resources Management, University of Patras, GR-30100 Agrinio, Greece
| | - Nikolina Charalampous
- Section of Animal Biology, Department of Biology, Faculty of Sciences, University of Patras, Rio, GR-26500 Patra, Greece
| | - Chara Kourkouta
- Section of Animal Biology, Department of Biology, Faculty of Sciences, University of Patras, Rio, GR-26500 Patra, Greece
| | - Stefanos Dailianis
- Section of Animal Biology, Department of Biology, Faculty of Sciences, University of Patras, Rio, GR-26500 Patra, Greece.
| |
Collapse
|
10
|
Abd El-Ghffar EA, El-Nashar HA, Eldahshan OA, Singab ANB. GC-MS analysis and hepatoprotective activity of the n-hexane extract of Acrocarpus fraxinifolius leaves against paracetamol-induced hepatotoxicity in male albino rats. PHARMACEUTICAL BIOLOGY 2017; 55:441-449. [PMID: 27937037 PMCID: PMC6130506 DOI: 10.1080/13880209.2016.1246575] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 05/27/2023]
Abstract
CONTEXT In Egypt, the burden of liver diseases is exceptionally high. OBJECTIVE To investigate the components of the n-hexane extract of Acrocarpus fraxinifolius Arn. (Leguminosae) and its hepatoprotective activity against paracetamol (APAP)-induced hepatotoxicity in rats. MATERIAL AND METHODS TRACE GC ultra gas chromatogaphic spectrometry was used for extract analysis. Thirty albino rats were divided into six groups (five rats in each). Group 1 was the healthy control; Groups 2 and 3 were healthy treated groups (250 and 500 mg/kg b.w. of the extract, respectively) for seven days. Group 4 was hepatotoxicity control (APAP intoxicated group). Groups 5 and 6 received APAP + extract 250 and APAP + extract 500, respectively. RESULTS Chromatographic analysis revealed the presence of 36 components. Major compounds were α-tocopherol (18.23%), labda-8 (20)-13-dien-15-oic acid (13.15%), lupeol (11.93%), phytol (10.95%) and squalene (7.19%). In the acute oral toxicity study, the mortality rates and behavioural signs of toxicity were zero in all groups (doses from 0 to 5 g/kg b.w. of A. fraxinifolius). LD50 was found to be greater than 5 g/kg of the extract. Only the high dose (500 mg/kg b.w.) of extract significantly alleviated the liver relative weight (4.01 ± 0.06) and biomarkers, as serum aspartate aminotransferase (62.87 ± 1.41), alanine aminotransferase (46.74 ± 1.45), alkaline phosphatase (65.96 ± 0.74), lipid profiles (180.39 ± 3.51), bilirubin profiles (2.30 ± 0.06) and hepatic lipid peroxidation (114.20 ± 2.06), and increased body weight (11.58 ± 0.20), serum protein profile (11.09 ± 0.46) and hepatic total antioxidant capacity (23.78 ± 0.66) in APAP-induced hepatotoxicity in rats. CONCLUSION Our study proves the antihepatotoxic/antioxidant efficacies of A. fraxinifolius hexane extract.
Collapse
Affiliation(s)
- Eman A. Abd El-Ghffar
- Department of Zoology, Faculty of Sciences, Ain Shams University, Abbassia, Cairo, Egypt
| | - Heba A.S. El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Omayma A. Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Abdel Nasser B. Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| |
Collapse
|
11
|
Theodosis-Nobelos P, Athanasekou C, Rekka EA. Dual antioxidant structures with potent anti-inflammatory, hypolipidemic and cytoprotective properties. Bioorg Med Chem Lett 2017; 27:4800-4804. [PMID: 29017787 DOI: 10.1016/j.bmcl.2017.09.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/20/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023]
Abstract
Novel amide derivatives of trolox, 3,5-di-tert-butyl-4-hydroxybenzoic acid, (E)-3-(3,5-di-tert-butyl-4-hydroxyphenyl)acrylic acid and cinnamic acid with cysteamine and l-cysteine ethyl ester were synthesised. In four cases, the disulfide derivatives were also isolated and tested. All compounds were examined for antioxidant activity, expressed as their ability to inhibit lipid peroxidation and to scavenge free radicals. They were found to demonstrate up to 17-fold better activity than that of the parent antioxidant acids. They could reduce acute inflammation up to 87%. The most active antioxidant compounds were further tested for their in vivo hypolipidemic effect, which ranged from 47% to 73%, and for their ability to protect the liver against oxidative toxicity caused by high paracetamol dose. The disulfide derivatives of 3,5-di-tert-butyl-4-hydroxybenzoic acid and cinnamic acid had no antioxidant activity and presented equal or lower anti-inflammatory effect than their thiol analogues, indicating that their molecular characteristics may not permit biological barrier penetration.
Collapse
Affiliation(s)
- Panagiotis Theodosis-Nobelos
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, Thessaloniki 54124, Greece
| | - Chrysoula Athanasekou
- Institute of Nanoscience & Nanotechnology, NCSR "Demokritos", 15310 Agia Paraskevi Attikis, Athens, Greece
| | - Eleni A Rekka
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, Thessaloniki 54124, Greece.
| |
Collapse
|
12
|
Wang X, Wu Q, Liu A, Anadón A, Rodríguez JL, Martínez-Larrañaga MR, Yuan Z, Martínez MA. Paracetamol: overdose-induced oxidative stress toxicity, metabolism, and protective effects of various compounds in vivo and in vitro. Drug Metab Rev 2017; 49:395-437. [PMID: 28766385 DOI: 10.1080/03602532.2017.1354014] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Paracetamol (APAP) is one of the most widely used and popular over-the-counter analgesic and antipyretic drugs in the world when used at therapeutic doses. APAP overdose can cause severe liver injury, liver necrosis and kidney damage in human beings and animals. Many studies indicate that oxidative stress is involved in the various toxicities associated with APAP, and various antioxidants were evaluated to investigate their protective roles against APAP-induced liver and kidney toxicities. To date, almost no review has addressed the APAP toxicity in relation to oxidative stress. This review updates the research conducted over the past decades into the production of reactive oxygen species (ROS), reactive nitrogen species (RNS), and oxidative stress as a result of APAP treatments, and ultimately their correlation with the toxicity and metabolism of APAP. The metabolism of APAP involves various CYP450 enzymes, through which oxidative stress might occur, and such metabolic factors are reviewed within. The therapeutics of a variety of compounds against APAP-induced organ damage based on their anti-oxidative effects is also discussed, in order to further understand the role of oxidative stress in APAP-induced toxicity. This review will throw new light on the critical roles of oxidative stress in APAP-induced toxicity, as well as on the contradictions and blind spots that still exist in the understanding of APAP toxicity, the cellular effects in terms of organ injury and cell signaling pathways, and finally strategies to help remedy such against oxidative damage.
Collapse
Affiliation(s)
- Xu Wang
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain.,b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Qinghua Wu
- c College of Life Science , Yangtze University , Jingzhou , China.,d Faculty of Informatics and Management , Center for Basic and Applied Research, University of Hradec Kralove , Hradec Kralove , Czech Republic
| | - Aimei Liu
- b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Arturo Anadón
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - José-Luis Rodríguez
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - María-Rosa Martínez-Larrañaga
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - Zonghui Yuan
- b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China.,e MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China.,f Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety , Wuhan , Hubei , China
| | - María-Aránzazu Martínez
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| |
Collapse
|
13
|
Leeming MG, Donald WA, O'Hair RAJ. Nontargeted Identification of Reactive Metabolite Protein Adducts. Anal Chem 2017; 89:5748-5756. [PMID: 28481086 DOI: 10.1021/acs.analchem.6b04604] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metabolic bioactivation of many different chemicals results in the formation of highly reactive compounds (chemically reactive metabolites, CRMs) that can lead to toxicity via binding to macromolecular targets (e.g., proteins or DNA). There is a need to develop robust, rapid, and nontargeted analytical techniques to determine the identity of the protein targets of CRMs and their sites of modification. Here, we introduce a nontargeted methodology capable of determining both the identity of a CRM formed from an administered compound as well as the protein targets modified by the reactive metabolite in a single experiment without prior information. Acetaminophen (N-acetyl-p-aminophenol, APAP) and 13C6-APAP were incubated with rat liver microsomes, which are known to bioactivate APAP to the reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI). Global tryptic digestion followed by liquid chromatographic/mass spectrometric (LC/MS) analysis was used to locate "twin" ion peaks of peptides adducted by NAPQI and for shotgun proteomics via tandem mass spectrometry (MS/MS). By the development of blended data analytics software called Xenophile, the identity of the amino acid residue that was adducted can be established, which eliminates the need for specific parametrization of protein database search algorithms. This combination of experimental design and data analysis software allows the identity of a CRM, the protein target, and the amino acid residues that are modified to be rapidly established directly from experimental data. Xenophile is freely available from https://github.com/mgleeming/Xenophile .
Collapse
Affiliation(s)
- Michael G Leeming
- School of Chemistry and Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne , Melbourne, Victoria 3010, Australia
| | - William A Donald
- School of Chemistry, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Richard A J O'Hair
- School of Chemistry and Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne , Melbourne, Victoria 3010, Australia
| |
Collapse
|
14
|
Rofaeil RR, Kamel MY, Abdelzaher WY. Different effects of selective β1-adrenoceptor antagonists, nebivolol or atenolol in acetaminophen-induced hepatotoxicity of rats. Fundam Clin Pharmacol 2017; 31:165-173. [DOI: 10.1111/fcp.12253] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/23/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Remon R. Rofaeil
- Faculty of Medicine; Minia University; Minia El Minia 61111 Egypt
| | - Maha Y. Kamel
- Faculty of Medicine; Minia University; Minia El Minia 61111 Egypt
| | | |
Collapse
|
15
|
Sabbioni G, Turesky RJ. Biomonitoring Human Albumin Adducts: The Past, the Present, and the Future. Chem Res Toxicol 2017; 30:332-366. [PMID: 27989119 PMCID: PMC5241710 DOI: 10.1021/acs.chemrestox.6b00366] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Indexed: 12/21/2022]
Abstract
Serum albumin (Alb) is the most abundant protein in blood plasma. Alb reacts with many carcinogens and/or their electrophilic metabolites. Studies conducted over 20 years ago showed that Alb forms adducts with the human carcinogens aflatoxin B1 and benzene, which were successfully used as biomarkers in molecular epidemiology studies designed to address the role of these chemicals in cancer risk. Alb forms adducts with many therapeutic drugs or their reactive metabolites such as β-lactam antibiotics, acetylsalicylic acid, acetaminophen, nonsteroidal anti-inflammatory drugs, chemotherapeutic agents, and antiretroviral therapy drugs. The identification and characterization of the adduct structures formed with Alb have served to understand the generation of reactive metabolites and to predict idiosyncratic drug reactions and toxicities. The reaction of candidate drugs with Alb is now exploited as part of the battery of screening tools to assess the potential toxicities of drugs. The use of gas chromatography-mass spectrometry, liquid chromatography, or liquid chromatography-mass spectrometry (LC-MS) enabled the identification and quantification of multiple types of Alb xenobiotic adducts in animals and humans during the past three decades. In this perspective, we highlight the history of Alb as a target protein for adduction to environmental and dietary genotoxicants, pesticides, and herbicides, common classes of medicinal drugs, and endogenous electrophiles, and the emerging analytical mass spectrometry technologies to identify Alb-toxicant adducts in humans.
Collapse
Affiliation(s)
- Gabriele Sabbioni
- Institute of Environmental and Occupational Toxicology, CH-6780 Airolo, Switzerland
- Alpine Institute of Chemistry and Toxicology, CH-6718 Olivone, Switzerland
- Walther-Straub-Institut für Pharmakologie
und Toxikologie, Ludwig-Maximilians-Universität München, D-80336 München, Germany
| | - Robert J. Turesky
- Masonic Cancer Center and Department of
Medicinal Chemistry, College of Pharmacy, University of Minnesota, 2231 Sixth Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
16
|
Wang J, Yang G, Dubrovsky AM, Choi J, Leung PSC. Xenobiotics and loss of tolerance in primary biliary cholangitis. World J Gastroenterol 2016; 22:338-348. [PMID: 26755880 PMCID: PMC4698496 DOI: 10.3748/wjg.v22.i1.338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/15/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023] Open
Abstract
Data from genome wide association studies and geoepidemiological studies established that a combination of genetic predisposition and environmental stimulation is required for the loss of tolerance in primary biliary cholangitis (PBC). The serologic hallmark of PBC are the presence of high titer anti-mitochondrial autoantibodies (AMA) that recognize the lipoyl domain of the mitochondrial pyruvate dehydrogenase E2 (PDC-E2) subunit. Extensive efforts have been directed to investigate the molecular basis of AMA. Recently, experimental data has pointed to the thesis that the breaking of tolerance to PDC-E2 is a pivotal event in the initial etiology of PBC, including environmental xenobiotics including those commonly found in cosmetics and food additives, suggesting that chemical modification of the PDC-E2 epitope may render its vulnerable to become a neo-antigen and trigger an immune response in genetically susceptible hosts. Here, we will discuss the natural history, genetics and immunobiology of PBC and structural constraints of PDC-E2 in AMA recognition which makes it vulnerable to chemical modification.
Collapse
|
17
|
Leeming MG, Gamon LF, Wille U, Donald WA, O'Hair RAJ. What Are the Potential Sites of Protein Arylation by N-Acetyl-p-benzoquinone Imine (NAPQI)? Chem Res Toxicol 2015; 28:2224-33. [PMID: 26523953 DOI: 10.1021/acs.chemrestox.5b00373] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Acetaminophen (paracetamol, APAP) is a safe and widely used analgesic medication when taken at therapeutic doses. However, APAP can cause potentially fatal hepatotoxicity when taken in overdose or in patients with metabolic irregularities. The production of the electrophilic and putatively toxic compound N-acetyl-p-benzoquinone imine (NAPQI), which cannot be efficiently detoxicated at high doses, is implicated in APAP toxicity. Numerous studies have identified that excess NAPQI can form covalent linkages to the thiol side chains of cysteine residues in proteins; however, the reactivity of NAPQI toward other amino acid side chains is largely unexplored. Here, we report a survey of the reactivity of NAPQI toward 11 N-acetyl amino acid methyl esters and four peptides. (1)H NMR analysis reveals that NAPQI forms covalent bonds to the side-chain functional groups of cysteine, methionine, tyrosine, and tryptophan residues. Analogous reaction products were observed when NAPQI was reacted with synthetic model peptides GAIL-X-GAILR for X = Cys, Met, Tyr, and Trp. Tandem mass spectrometry peptide sequencing showed that the NAPQI modification sites are located on the "X" residue in each case. However, when APAP and the GAIL-X-GAILR peptide were incubated with rat liver microsomes that contain many metabolic enzymes, NAPQI formed by oxidative metabolism reacted with GAIL-C-GAILR exclusively. For the peptides where X = Met, Tyr, and Trp, competing reactions between NAPQI and alternative nucleophiles precluded arylation of the target peptide by NAPQI. Although Cys residues are favorably targeted under these conditions, these data suggest that NAPQI can, in principle, also damage proteins at Met, Tyr, and Trp residues.
Collapse
Affiliation(s)
- Michael G Leeming
- School of Chemistry and ARC Centre of Excellence for Free Radical Chemistry and Biotechnology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne , Parkville, Victoria 3010, Australia
| | - Luke F Gamon
- School of Chemistry and ARC Centre of Excellence for Free Radical Chemistry and Biotechnology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne , Parkville, Victoria 3010, Australia
| | - Uta Wille
- School of Chemistry and ARC Centre of Excellence for Free Radical Chemistry and Biotechnology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne , Parkville, Victoria 3010, Australia
| | - William A Donald
- School of Chemistry, University of New South Wales , Sydney, NSW 2052, Australia
| | - Richard A J O'Hair
- School of Chemistry and ARC Centre of Excellence for Free Radical Chemistry and Biotechnology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne , Parkville, Victoria 3010, Australia
| |
Collapse
|
18
|
Zhou X, Cheung CM, Yang JM, Or PMY, Lee WYW, Yeung JHK. Danshen (Salvia miltiorrhiza) water extract inhibits paracetamol-induced toxicity in primary rat hepatocytes via reducing CYP2E1 activity and oxidative stress. J Pharm Pharmacol 2015; 67:980-989. [PMID: 25645193 DOI: 10.1111/jphp.12381] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/07/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This study aimed to investigate the protective effects of Danshen (Salvia miltiorrhiza) water extract (DSE) and its major phenolic acid components against CYP2E1-mediated paracetamol (APAP)-induced hepatic toxicity. METHODS The protection and underlying mechanisms were detected in CYP2E1 overexpression primary rat hepatocytes by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, alamar blue assay, CYP2E1 inhibition assay and glutathione assay. KEY FINDINGS After APAP treatment, DSE (0.06-1 mg/ml) significantly increased cell viability in MTT assay. Two major components danshensu (8.2-130.5 μm) and salvianolic acid B (Sal B; 3.3-53.5 μm) mainly contributed to this protection, but rosmarinic acid, protocatechuic aldehyde and Sal A did not. Alamar blue assay showed that DSE, danshensu and Sal B maintained mitochondrial metabolic activity. DSE inhibited CYP2E1 (Ki = 1.46 mg/ml) in a mixed mode in rat liver microsomes in vitro; DSE decreased APAP-induced total glutathione depletion and preserved redox status (GSH/GSSG ratio) in hepatocytes. Danshensu and Sal B did not inhibit CYP2E1 or decrease total glutathione depletion, but preserved redox status. CONCLUSIONS DSE protected hepatocytes against APAP-induced injury via maintenance of mitochondrial metabolic activity, CYP2E1 inhibition, reduction of total glutathione depletion and preservation of redox status. Danshensu and Sal B were mainly responsible for this protection.
Collapse
Affiliation(s)
- Xuelin Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ching Mei Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jia-Ming Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Penelope M Y Or
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wayne Y W Lee
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - John H K Yeung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Aminotriazole alleviates acetaminophen poisoning via downregulating P450 2E1 and suppressing inflammation. PLoS One 2015; 10:e0122781. [PMID: 25884831 PMCID: PMC4401561 DOI: 10.1371/journal.pone.0122781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/14/2015] [Indexed: 12/13/2022] Open
Abstract
Aminotriazole (ATZ) is commonly used as a catalase (CAT) inhibitor. We previously found ATZ attenuated oxidative liver injury, but the underlying mechanisms remain unknown. Acetaminophen (APAP) overdose frequently induces life-threatening oxidative hepatitis. In the present study, the potential hepatoprotective effects of ATZ on oxidative liver injury and the underlying mechanisms were further investigated in a mouse model with APAP poisoning. The experimental data indicated that pretreatment with ATZ dose- and time-dependently suppressed the elevation of plasma aminotransferases in APAP exposed mice, these effects were accompanied with alleviated histological abnormality and improved survival rate of APAP-challenged mice. In mice exposed to APAP, ATZ pretreatment decreased the CAT activities, hydrogen peroxide (H2O2) levels, malondialdehyde (MDA) contents, myeloperoxidase (MPO) levels in liver and reduced TNF-α levels in plasma. Pretreatment with ATZ also downregulated APAP-induced cytochrome P450 2E1 (CYP2E1) expression and JNK phosphorylation. In addition, posttreatment with ATZ after APAP challenge decreased the levels of plasma aminotransferases and increased the survival rate of experimental animals. Posttreatment with ATZ had no effects on CYP2E1 expression or JNK phosphorylation, but it significantly decreased the levels of plasma TNF-α. Our data indicated that the LD50 of ATZ in mice was 5367.4 mg/kg body weight, which is much higher than the therapeutic dose of ATZ in the present study. These data suggested that ATZ might be effective and safe in protect mice against APAP-induced hepatotoxicity, the beneficial effects might resulted from downregulation of CYP2E1 and inhibiton of inflammation.
Collapse
|
20
|
Ghanem CI, Rudraiah S, Bataille AM, Vigo MB, Goedken MJ, Manautou JE. Role of nuclear factor-erythroid 2-related factor 2 (Nrf2) in the transcriptional regulation of brain ABC transporters during acute acetaminophen (APAP) intoxication in mice. Biochem Pharmacol 2015; 94:203-211. [PMID: 25667042 DOI: 10.1016/j.bcp.2015.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED Changes in expression of liver ABC transporters have been described during acute APAP intoxication. However, the effect of APAP on brain ABC transporters is poorly understood. The aim of this study was to evaluate the effect of APAP on brain ABC transporters expression and the role of the oxidative stress sensor Nrf2. Male C57BL/6J mice were administered APAP (400mg/kg) for analysis of brain mRNA and protein expression of Mrp1-6, Bcrp and P-gp. The results show induction of P-gp, Mrp2 and Mrp4 proteins, with no changes in Bcrp, Mrp1 or Mrp5-6. The protein values were accompanied by corresponding changes in mRNA levels. Additionally, brain Nrf2 nuclear translocation and expression of two Nrf2 target genes, NAD(P)H quinone oxidoreductase 1 (Nqo1) and Hemoxygenase 1 (Ho-1), was evaluated at 6, 12 and 24h after APAP treatment. Nrf2 nuclear content increased by 58% at 12h after APAP along with significant increments in mRNA and protein expression of Nqo1 and Ho-1. Furthermore, APAP treated Nrf2 knockout mice did not increase mRNA or protein expression of Mrp2 and Mrp4 as observed in wildtypes. In contrast, P-gp induction by APAP was observed in both genotypes. In conclusion, acute APAP intoxication induces protein expression of brain P-gp, Mrp2 and Mrp4. This study also suggests that brain changes in Mrp2 and Mrp4 expression may be due to in situ Nrf2 activation by APAP, while P-gp induction is independent of Nrf2 function. The functional consequences of these changes in brain ABC transporters by APAP deserve further attention.
Collapse
Affiliation(s)
- Carolina I Ghanem
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina; Cátedra de Fisiopatología. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Amy M Bataille
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - María B Vigo
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Michael J Goedken
- Office of Translational Science, Rutgers University, Piscataway, NJ 08854, United States
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States.
| |
Collapse
|
21
|
Ware BR, Berger DR, Khetani SR. Prediction of Drug-Induced Liver Injury in Micropatterned Co-cultures Containing iPSC-Derived Human Hepatocytes. Toxicol Sci 2015; 145:252-62. [PMID: 25716675 DOI: 10.1093/toxsci/kfv048] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Primary human hepatocytes (PHHs) are a limited resource for drug screening, their quality for in vitro use can vary considerably across different lots, and a lack of available donor diversity restricts our understanding of how human genetics affect drug-induced liver injury (DILI). Induced pluripotent stem cell-derived human hepatocyte-like cells (iPSC-HHs) could provide a complementary tool to PHHs for high-throughput drug screening, and ultimately enable personalized medicine. Here, we hypothesized that previously developed iPSC-HH-based micropatterned co-cultures (iMPCCs) with murine embryonic fibroblasts could be amenable to long-term drug toxicity assessment. iMPCCs, created in industry-standard 96-well plates, were treated for 6 days with a set of 47 drugs, and multiple functional endpoints (albumin, urea, ATP) were evaluated in dosed cultures against vehicle-only controls to enable binary toxicity decisions. We found that iMPCCs correctly classified 24 of 37 hepatotoxic drugs (65% sensitivity), while all 10 non-toxic drugs tested were classified as such in iMPCCs (100% specificity). On the other hand, conventional confluent cultures of iPSC-HHs failed to detect several liver toxins that were picked up in iMPCCs. Results for DILI detection in iMPCCs were remarkably similar to published data in PHH-MPCCs (65% versus 70% sensitivity) that were dosed with the same drugs. Furthermore, iMPCCs detected the relative hepatotoxicity of structural drug analogs and recapitulated known mechanisms of acetaminophen toxicity in vitro. In conclusion, iMPCCs could provide a robust tool to screen for DILI potential of large compound libraries in early stages of drug development using an abundant supply of commercially available iPSC-HHs.
Collapse
Affiliation(s)
- Brenton R Ware
- *School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523 and Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523
| | - Dustin R Berger
- *School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523 and Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523
| | - Salman R Khetani
- *School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523 and Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523 *School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523 and Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
22
|
Schattenberg JM, Czaja MJ. Regulation of the effects of CYP2E1-induced oxidative stress by JNK signaling. Redox Biol 2014; 3:7-15. [PMID: 25462060 PMCID: PMC4218941 DOI: 10.1016/j.redox.2014.09.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 02/07/2023] Open
Abstract
The generation of excessive amounts of reactive oxygen species (ROS) leads to cellular oxidative stress that underlies a variety of forms of hepatocyte injury and death including that from alcohol. Although ROS can induce cell damage through direct effects on cellular macromolecules, the injurious effects of ROS are mediated largely through changes in signal transduction pathways such as the mitogen-activated protein kinase c-Jun N-terminal kinase (JNK). In response to alcohol, hepatocytes have increased levels of the enzyme cytochrome P450 2E1 (CYP2E1) which generates an oxidant stress that promotes the development of alcoholic steatosis and liver injury. These effects are mediated in large part through overactivation of JNK that alters cell death pathways. Targeting the JNK pathway or its downstream effectors may be a useful therapeutic approach to the oxidative stress generated by CYP2E1 in alcoholic liver disease.
Collapse
Affiliation(s)
- Jörn M Schattenberg
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Mark J Czaja
- Department of Medicine and Marion Bessin Liver Research Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
23
|
Gosselin S, Juurlink DN, Kielstein JT, Ghannoum M, Lavergne V, Nolin TD, Hoffman RS. Extracorporeal treatment for acetaminophen poisoning: recommendations from the EXTRIP workgroup. Clin Toxicol (Phila) 2014; 52:856-67. [PMID: 25133498 DOI: 10.3109/15563650.2014.946994] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The Extracorporeal Treatments in Poisoning (EXTRIP) workgroup was created to provide evidence-based recommendations on the use of extracorporeal treatments (ECTR) in poisoning and the results are presented here for acetaminophen (APAP). METHODS After a systematic review of the literature, a subgroup selected and reviewed the articles and summarized clinical and toxicokinetic data in order to propose structured voting statements following a pre-determined format. A two-round modified Delphi method was chosen to reach a consensus on voting statements, and the RAND/UCLA Appropriateness Method was used to quantify disagreement. Following discussion, a second vote determined the final recommendations. RESULTS Twenty-four articles (1 randomized controlled trial, 1 observational study, 2 pharmacokinetic studies, and 20 case reports or case series) were identified, yielding an overall very low quality of evidence for all recommendations. Clinical data on 135 patients and toxicokinetic data on 54 patients were analyzed. Twenty-three fatalities were reviewed. The workgroup agreed that N-acetylcysteine (NAC) is the mainstay of treatment, and that ECTR is not warranted in most cases of APAP poisoning. However, given that APAP is dialyzable, the workgroup agreed that ECTR is suggested in patients with excessively large overdoses who display features of mitochondrial dysfunction. This is reflected by early development of altered mental status and severe metabolic acidosis prior to the onset of hepatic failure. Specific recommendations for ECTR include an APAP concentration over 1000 mg/L if NAC is not administered (1D), signs of mitochondrial dysfunction and an APAP concentration over 700 mg/L (4630 mmol/L) if NAC is not administered (1D) and signs of mitochondrial dysfunction and an APAP concentration over 900 mg/L (5960 mmol/L) if NAC is administered (1D). Intermittent hemodialysis (HD) is the preferred ECTR modality in APAP poisoning (1D). CONCLUSION APAP is amenable to extracorporeal removal. Due to the efficacy of NAC, ECTR is reserved for rare situations when the efficacy of NAC has not been definitively demonstrated.
Collapse
Affiliation(s)
- S Gosselin
- Department of Emergency Medicine, Medical Toxicology Service, McGill University Health Centre, McGill University , Montréal, QC , Canada
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Ukairo O, McVay M, Krzyzewski S, Aoyama S, Rose K, Andersen ME, Khetani SR, Lecluyse EL. Bioactivation and toxicity of acetaminophen in a rat hepatocyte micropatterned coculture system. J Biochem Mol Toxicol 2013; 27:471-8. [PMID: 23918466 DOI: 10.1002/jbt.21512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/16/2013] [Accepted: 06/28/2013] [Indexed: 12/13/2022]
Abstract
We have recently shown that primary rat hepatocytes organized in micropatterned cocultures with murine embryonic fibroblasts (HepatoPac™) maintain high levels of liver functions for at least 4 weeks. In this study, rat HepatoPac was assessed for its utility to study chemical bioactivation and associated hepatocellular toxicity. Treatment of HepatoPac cultures with acetaminophen (APAP) over a range of concentrations (0-15 mM) was initiated at 1, 2, 3, or 4 weeks followed by the assessment of morphological and functional endpoints. Consistent and reproducible concentration-dependent effects on hepatocyte structure, viability, and basic functions were observed over the 4-week period, and were exacerbated by depleting glutathione using buthionine sulfoximine or inducing CYP3A using dexamethasone, presumably due to increased reactive metabolite-induced stress and adduct formation. In conclusion, the results from this study demonstrate that rat HepatoPac represents a structurally and functionally stable hepatic model system to assess the long-term effects of bioactivated compounds.
Collapse
|
25
|
Environment and primary biliary cirrhosis: electrophilic drugs and the induction of AMA. J Autoimmun 2013; 41:79-86. [PMID: 23352659 DOI: 10.1016/j.jaut.2012.12.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 12/12/2022]
Abstract
Environmental stimulation is a major factor in the initiation and perpetuation of autoimmune diseases. We have addressed this issue and focused on primary biliary cirrhosis (PBC), an autoimmune disease of the liver. Immunologically, PBC is distinguished by immune mediated destruction of the intra hepatic bile ducts and the presence of high titer antimitochondrial autoantibodies (AMA) directed against a highly specific epitope within the lipoic acid binding domain of the pyruvate dehydrogenase E2 subunit (PDC-E2). We submit that the uniqueness of AMA epitope specificity and the conformational changes of the PDC-E2 lipoyl domain during physiological acyl transfer could be the lynchpin to the etiology of PBC and postulate that chemical xenobiotics modification of the lipoyl domain of PDC-E2 is sufficient to break self-tolerance, with subsequent production of AMA in patients with PBC. Indeed, using quantitative structure activity relationship (QSAR) analysis on a peptide-xenobiotic conjugate microarray platform, we have demonstrated that when the lipoyl domain of PDC-E2 was modified with specific synthetic small molecule lipoyl mimics, the ensuing structures displayed highly specific reactivity to PBC sera, at levels often higher than the native PDC-E2 molecule. Hereby, we discuss our recent QSAR analysis data on specific AMA reactivity against a focused panel of lipoic acid mimic in which the lipoyl di-sulfide bond are modified. Furthermore, data on the immunological characterization of antigen and Ig isotype specificities against one such lipoic acid mimic; 6,8-bis(acetylthio)octanoic acid (SAc), when compared with rPDC-E2, strongly support a xenobiotic etiology in PBC. This observation is of particular significance in that approximately one third of patients who have taken excessive acetaminophen (APAP) developed AMA with same specificity as patients with PBC, suggesting that the lipoic domain are a target of APAP electrophilic metabolites such as NAPQI. We submit that in genetically susceptible hosts, electrophilic modification of lipoic acid in PDC-E2 by acetaminophen or similar drugs can facilitate loss of tolerance and lead to the development of PBC.
Collapse
|
26
|
Kang LI, Mars WM, Michalopoulos GK. Signals and cells involved in regulating liver regeneration. Cells 2012; 1:1261-1292. [PMID: 24710554 PMCID: PMC3901148 DOI: 10.3390/cells1041261] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/27/2012] [Accepted: 12/07/2012] [Indexed: 12/11/2022] Open
Abstract
Liver regeneration is a complex phenomenon aimed at maintaining a constant liver mass in the event of injury resulting in loss of hepatic parenchyma. Partial hepatectomy is followed by a series of events involving multiple signaling pathways controlled by mitogenic growth factors (HGF, EGF) and their receptors (MET and EGFR). In addition multiple cytokines and other signaling molecules contribute to the orchestration of a signal which drives hepatocytes into DNA synthesis. The other cell types of the liver receive and transmit to hepatocytes complex signals so that, in the end of the regenerative process, complete hepatic tissue is assembled and regeneration is terminated at the proper time and at the right liver size. If hepatocytes fail to participate in this process, the biliary compartment is mobilized to generate populations of progenitor cells which transdifferentiate into hepatocytes and restore liver size.
Collapse
Affiliation(s)
- Liang-I Kang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
27
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
28
|
Saritas A, Kandis H, Baltaci D, Yildirim U, Kaya H, Karakus A, Colakoglu S, Memisogullari R, Kara IH. N-Acetyl cysteine and erdosteine treatment in acetaminophen-induced liver damage. Toxicol Ind Health 2012; 30:670-8. [PMID: 23070635 DOI: 10.1177/0748233712463780] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE This study is aimed to investigate the efficacy of erdosteine usage in acetaminophen-induced liver damage and to compare it with N-acetyl cysteine (NAC) in the treatment and prevention of liver toxicity due to overdose of acetaminophen. METHODS The rats were separated into the following six groups of seven rats each: control group; acetaminophen (1 g/kg, orally); acetaminophen (1 g/kg, orally) + erdosteine (150 mg/kg/day, orally); acetaminophen (1 g/kg, orally) + NAC (140 mg/kg loading dose, followed by 70 mg/kg, orally); NAC (140 mg/kg loading dose, followed by 70 mg/kg, orally); erdosteine (150 mg/kg/kg, orally), subsequently. In all the groups, potential liver injuries were evaluated using biochemical and hematological analyses, oxidant-antioxidant parameters and histopathological parameters. RESULTS In acetaminophen-treated group, levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total oxidant status (TOS) in the blood, prothrombin time (PT) and international normalized ratio (INR) were significantly increased when compared with controls. However, total antioxidant capacity (TAC) and glutathione (GSH) levels were decreased in group treated with acetaminophen, when compared with control group. Levels of AST, ALT and TOS, PT and INR were decreased in groups treated with NAC and erdosteine after acetaminophen administration, but the levels of TAC and GSH were increased. Histopathological improvements were observed in the groups treated with NAC and erdosteine after acetaminophen administration. CONCLUSION The present study demonstrated that, in the prevention of liver damage induced by acetaminophen intoxication, an early treatment with a single dose of erdosteine was beneficial instead of NAC administration.
Collapse
Affiliation(s)
- Ayhan Saritas
- Department of Emergency Medicine, Duzce University School of Medicine, Duzce, Turkey
| | - Hayati Kandis
- Department of Emergency Medicine, Duzce University School of Medicine, Duzce, Turkey
| | - Davut Baltaci
- Department of Family Medicine, Duzce University School of Medicine, Duzce, Turkey
| | - Umran Yildirim
- Department of Pathology, Duzce University School of Medicine, Duzce, Turkey
| | - Halil Kaya
- Department of Emergency Medicine, Harran University School of Medicine, Sanliurfa, Turkey
| | - Ali Karakus
- Department of Emergency Medicine, Mustafa Kemal University School of Medicine, Hatay, Turkey
| | - Serdar Colakoglu
- Department of Anatomy, Duzce University School of Medicine, Duzce, Turkey
| | | | - Ismail Hamdi Kara
- Department of Family Medicine, Duzce University School of Medicine, Duzce, Turkey
| |
Collapse
|
29
|
Slater D, Kunnathil S, McBride J, Koppala R. Pharmacology of nonsteroidal antiinflammatory drugs and opioids. Semin Intervent Radiol 2012; 27:400-11. [PMID: 22550382 DOI: 10.1055/s-0030-1267855] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic pain affects up to 50 million Americans every day. Traditional treatment has included acetaminophen, nonsteroidal antiinflammatory drugs (NSAIDs), or opioids. The combination of NSAIDs and opioids can provide effective treatment for up to 90% of patients with chronic pain, but the NSAIDs have the potential for significant, even life-threatening side effects. Additionally, the nonselective cyclooxygenase inhibitors with 16,000 deaths per year in the United States might not be any safer. The opioids are great for short-term pain, but may need to be adjusted or changed frequently due to the development of tolerance. Understanding of the mechanism of opioids and NSAIDs has improved greatly over the past decade, but is still incomplete.
Collapse
Affiliation(s)
- Dick Slater
- Department of Radiology, University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | |
Collapse
|
30
|
Leung PSC, Lam K, Kurth MJ, Coppel RL, Gershwin ME. Xenobiotics and autoimmunity: does acetaminophen cause primary biliary cirrhosis? Trends Mol Med 2012; 18:577-82. [PMID: 22920894 DOI: 10.1016/j.molmed.2012.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/08/2012] [Accepted: 07/11/2012] [Indexed: 01/09/2023]
Abstract
The serologic hallmark of primary biliary cirrhosis (PBC) is the presence of antimitochondrial autoantibodies (AMAs) directed against the E2 subunit of the pyruvate dehydrogenase complex (PDC-E2). The PBC-related autoepitope of PDC-E2 contains lipoic acid, and previous work has demonstrated that mimics of lipoic acid following immunization of mice lead to a PBC-like disease. Furthermore, approximately one-third of patients who have ingested excessive amounts of acetaminophen (paracetamol) develop AMA of the same specificity as patients with PBC. Quantitative structure-activity relationship (QSAR) data indicates that acetaminophen metabolites are particularly immunoreactive with AMA, and we submit that in genetically susceptible hosts, electrophilic modification of lipoic acid in PDC-E2 by acetaminophen or similar drugs can facilitate a loss of tolerance and lead to the development of PBC.
Collapse
Affiliation(s)
- Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|
31
|
Teng CY, Lai YL, Huang HI, Hsu WH, Yang CC, Kuo WH. Tournefortia sarmentosa extract attenuates acetaminophen-induced hepatotoxicity. PHARMACEUTICAL BIOLOGY 2012; 50:291-396. [PMID: 22085220 DOI: 10.3109/13880209.2011.602695] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
CONTEXT Tournefortia sarmentosa Lam. (Boraginaceae), a Chinese herbal medicine, is commonly used as a detoxicant or anti-inflammatory agent. OBJECTIVE As acetaminophen (APAP) is a well-known hepatotoxin, we investigated the effect of the aqueous extract of the T. sarmentosa on APAP-induced hepatotoxicity in vivo and in vitro. MATERIALS AND METHODS Levels of liver function markers serum glutamate oxaloacetate transaminase (SGOT), glutamate pyruvate transaminase (SGPT), and alkaline phosphatase (ALP), inflammatory markers tumor necrosis factor (TNF)-α, interleukin (IL)-1b, and IL-6 in serum, and antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx), as well as lipid peroxidation were determined. RESULTS T. sarmentosa significantly reduced the elevated liver function (SGOT, SGPT, and ALP, p < 0.01) and inflammatory markers (TNF-α, IL-1β, and IL-6, p < 0.01) in serum of APAP-intoxicated rats. Malondialdehyde level (p < 0.05) and antioxidant enzyme levels (CAT, SOD, and GPx, p < 0.05) were also reduced in APAP-intoxicated rats treated with T. sarmentosa. Incubation of rat hepatocyte cell line clone-9 cells with APAP reduced cell viability and increased the extent of lipid peroxidation. APAP stimulation also reduced the level of glutathione (GSH) and caused reduction in the activities of the antioxidant enzymes, CAT, SOD, and GPx. Pretreatment of hepatocytes with T. sarmentosa aqueous extract before and during APAP stimulation attenuated the extent of lipid peroxidation, increased cell viability and GSH level, and enhanced the activities of antioxidant enzymes. DISCUSSION AND CONCLUSION These data suggest that the aqueous extract of T. sarmentosa can prevent APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Chun-Yuh Teng
- Division of Gastroenterology, Department of Internal Medicine, Armed-Forces Taichung General Hospital, Taiping City, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Toyoda Y, Tamai M, Kashikura K, Kobayashi S, Fujiyama Y, Soga T, Tagawa YI. Acetaminophen-induced hepatotoxicity in a liver tissue model consisting of primary hepatocytes assembling around an endothelial cell network. Drug Metab Dispos 2012; 40:169-77. [PMID: 22010217 DOI: 10.1124/dmd.111.041137] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Primary hepatocytes have been used in drug development for the evaluation of hepatotoxicity of candidate compounds. However, the rapid depression of their hepatic characters in vitro must be improved to predict toxicity with higher accuracy. We have hypothesized that a well organized tissue construct that includes nonparenchymal cells and appropriate scaffold material(s) could overcome this difficulty by remediating the viability and physiological function of primary hepatocytes. In this study, we constructed an in vitro liver tissue model, consisting of mouse primary hepatocytes assembling around an endothelial cell network on Engelbreth-Holm-Swarm gel, and examined its response to acetaminophen treatment. The increase in lactate dehydrogenase release after the exposure to acetaminophen was induced earlier in the liver tissue model than in monolayer hepatocytes alone, suggesting that the tissue model was more sensitive to an acetaminophen-induced toxicity. On the basis of our results, we conclude that liver tissue models of this kind may enhance the responses of hepatocytes against xenobiotics via the maintenance of hepatic genes and functions such as cytochrome P450s. These findings will contribute to the development of more accurate systems for evaluating hepatotoxicity.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B-51 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Buchard A, Eefsen M, Semb S, Andersen SE, Morling N, Bendtsen F, Larsen FS, Dalhoff K. The role of the glutathione S-transferase genes GSTT1, GSTM1, and GSTP1 in acetaminophen-poisoned patients. Clin Toxicol (Phila) 2011; 50:27-33. [DOI: 10.3109/15563650.2011.639713] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
34
|
Ellison CM, Enoch SJ, Cronin MTD. A review of the use ofin silicomethods to predict the chemistry of molecular initiating events related to drug toxicity. Expert Opin Drug Metab Toxicol 2011; 7:1481-95. [DOI: 10.1517/17425255.2011.629186] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
35
|
Pickering G, Schneider E, Papet I, Pujos-Guillot E, Pereira B, Simen E, Dubray C, Schoeffler P. Acetaminophen metabolism after major surgery: a greater challenge with increasing age. Clin Pharmacol Ther 2011; 90:707-11. [PMID: 21975347 DOI: 10.1038/clpt.2011.176] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Patients undergoing major surgery represent a good model for the study of the hepatic metabolism of acetaminophen (APAP) after surgery and for the evaluation of how the detoxification process is influenced by aging. Thirty patients received intravenous APAP (1 g/6 h) for 4 days (D1-D4). Daily 24-h urinary metabolites-cysteine-APAP, mercapturate-APAP, APAP, and glucuronide and sulfate conjugates-as well as blood glutathione levels were compared with repeated-measures analysis of variance (significance, P<0.05). Between D1 and D4, cysteine-APAP increased (308±308 mg vs. 570±512 mg, P=0.005), and sulfate and glucuronide conjugates decreased (1,365±1,084 mg vs. 694±600 mg, P<0.0001 and 2,418±817 mg vs. 1,513±1,076 mg, P=0.011, respectively). Blood glutathione decreased (790±125 vs. 623±132 µmol/l, P<0.0001. These changes increased with aging. APAP disposition after major surgery shifts toward the oxidative pathways of metabolism, and this is enhanced with aging. Supplementation with sulfur-containing amino acids should be investigated further as it might minimize the effect on antioxidant defenses, especially in older persons undergoing more extensive surgical procedures.
Collapse
Affiliation(s)
- G Pickering
- CHU Clermont-Ferrand, Centre de Pharmacologie Clinique, INSERM, CIC 501, UMR 766, Laboratoire de Pharmacologie, Faculté de Médecine, Clermont Université, Clermont-Ferrand, France.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Naiyanetr P, Butler JD, Meng L, Pfeiff J, Kenny TP, Guggenheim KG, Reiger R, Lam K, Kurth MJ, Ansari AA, Coppel RL, López-Hoyos M, Gershwin ME, Leung PSC. Electrophile-modified lipoic derivatives of PDC-E2 elicits anti-mitochondrial antibody reactivity. J Autoimmun 2011; 37:209-16. [PMID: 21763105 DOI: 10.1016/j.jaut.2011.06.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/24/2011] [Accepted: 06/01/2011] [Indexed: 12/27/2022]
Abstract
Our laboratory has hypothesized that xenobiotic modification of the native lipoyl moiety of the major mitochondrial autoantigen, the E2 subunit of the pyruvate dehydrogenase complex (PDC-E2), may lead to loss of self-tolerance in primary biliary cirrhosis (PBC). This thesis is based on the finding of readily detectable levels of immunoreactivity of PBC sera against extensive panels of protein microarrays containing mimics of the inner lipoyl domain of PDC-E2 and subsequent quantitative structure-activity relationships (QSARs). Importantly, we have demonstrated that murine immunization with one such mimic, 2-octynoic acid coupled to bovine serum albumin (BSA), induces anti-mitochondrial antibodies (AMAs) and cholangitis. Based upon these data, we have focused on covalent modifications of the lipoic acid disulfide ring and subsequent analysis of such xenobiotics coupled to a 15mer of PDC-E2 for immunoreactivity against a broad panel of sera from patients with PBC and controls. Our results demonstrate that AMA-positive PBC sera demonstrate marked reactivity against 6,8-bis(acetylthio)octanoic acid, implying that chemical modification of the lipoyl ring, i.e. disruption of the S-S disulfide, renders lipoic acid to its reduced form that will promote xenobiotic modification. This observation is particularly significant in light of the function of the lipoyl moiety in electron transport of which the catalytic disulfide constantly opens and closes and, thus, raises the intriguing thesis that common electrophilic agents, i.e. acetaminophen or non-steroidal anti-inflammatory drugs (NSAIDs), may lead to xenobiotic modification in genetically susceptible individuals that results in the generation of AMAs and ultimately clinical PBC.
Collapse
Affiliation(s)
- Phornnop Naiyanetr
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
McLachlan AJ, Bath S, Naganathan V, Hilmer SN, Le Couteur DG, Gibson SJ, Blyth FM. Clinical pharmacology of analgesic medicines in older people: impact of frailty and cognitive impairment. Br J Clin Pharmacol 2011; 71:351-64. [PMID: 21284694 DOI: 10.1111/j.1365-2125.2010.03847.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Pain is highly prevalent in frail older people who often have multiple co-morbidities and multiple medicines. Rational prescribing of analgesics in frail older people is complex due to heterogeneity in drug disposition, comorbid medical conditions, polypharmacy and variability in analgesic response in this population. A critical issue in managing older people with pain is the need for judicious choice of analgesics based on a comprehensive medical and medication history. Care is needed in the selection of analgesic medicine to avoid drug-drug or drug-disease interactions. People living with dementia and cognitive impairment have suboptimal pain relief which in part may be related to altered pharmacodynamics of analgesics and challenges in the systematic assessment of pain intensity in this patient group. In the absence of rigorously controlled trials in frail older people and those with cognitive impairment a pharmacologically-guided approach can be used to optimize pain management which requires a systematic understanding of the pharmacokinetics and pharmacodynamics of analgesics in frail older people with or without changes in cognition.
Collapse
Affiliation(s)
- Andrew J McLachlan
- Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia.
| | | | | | | | | | | | | |
Collapse
|
38
|
Begriche K, Massart J, Robin MA, Borgne-Sanchez A, Fromenty B. Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. J Hepatol 2011; 54:773-94. [PMID: 21145849 DOI: 10.1016/j.jhep.2010.11.006] [Citation(s) in RCA: 378] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 11/05/2010] [Accepted: 11/09/2010] [Indexed: 02/08/2023]
Abstract
Numerous investigations have shown that mitochondrial dysfunction is a major mechanism of drug-induced liver injury, which involves the parent drug or a reactive metabolite generated through cytochromes P450. Depending of their nature and their severity, the mitochondrial alterations are able to induce mild to fulminant hepatic cytolysis and steatosis (lipid accumulation), which can have different clinical and pathological features. Microvesicular steatosis, a potentially severe liver lesion usually associated with liver failure and profound hypoglycemia, is due to a major inhibition of mitochondrial fatty acid oxidation (FAO). Macrovacuolar steatosis, a relatively benign liver lesion in the short term, can be induced not only by a moderate reduction of mitochondrial FAO but also by an increased hepatic de novo lipid synthesis and a decreased secretion of VLDL-associated triglycerides. Moreover, recent investigations suggest that some drugs could favor lipid deposition in the liver through primary alterations of white adipose tissue (WAT) homeostasis. If the treatment is not interrupted, steatosis can evolve toward steatohepatitis, which is characterized not only by lipid accumulation but also by necroinflammation and fibrosis. Although the mechanisms involved in this aggravation are not fully characterized, it appears that overproduction of reactive oxygen species by the damaged mitochondria could play a salient role. Numerous factors could favor drug-induced mitochondrial and metabolic toxicity, such as the structure of the parent molecule, genetic predispositions (in particular those involving mitochondrial enzymes), alcohol intoxication, hepatitis virus C infection, and obesity. In obese and diabetic patients, some drugs may induce acute liver injury more frequently while others may worsen the pre-existent steatosis (or steatohepatitis).
Collapse
Affiliation(s)
- Karima Begriche
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, USA
| | | | | | | | | |
Collapse
|
39
|
Development of an acetaminophen amperometric biosensor based on peroxidase entrapped in polyacrylamide microgels. Biosens Bioelectron 2011; 26:1883-9. [DOI: 10.1016/j.bios.2010.03.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 03/18/2010] [Accepted: 03/18/2010] [Indexed: 11/20/2022]
|
40
|
Abstract
Paracetamol (acetaminophen) is a worldwide used analgesic and antipyretic drug. It is metabolised via several metabolic pathways, including glucuronidation, sulfation, oxidation, hydroxylation, and deacetylation: Hepatic and other organ damage may occur, especially in overdose, because of the accumulation of a toxic metabolite. Intersubject and ethnic differences have been reported in paracetamol metabolism activation, suggesting possible differences in susceptibility to toxicity and in pain alleviation, linked to different pharmacogenetic profiles. This article aims at reviewing, in the literature, the links between paracetamol metabolism and enzyme genotypes in the context of toxic side effects and efficacy of paracetamol in therapeutics.
Collapse
Affiliation(s)
- Lizi Zhao
- Institute of Clinical Pharmacology, Sun Yat-Sen University, Guangzhou, China
| | | |
Collapse
|
41
|
Effects on feeding rate and biomarker responses of marine mussels experimentally exposed to propranolol and acetaminophen. Anal Bioanal Chem 2009; 396:649-56. [DOI: 10.1007/s00216-009-3182-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/16/2009] [Accepted: 09/21/2009] [Indexed: 10/20/2022]
|
42
|
Copple IM, Goldring CE, Jenkins RE, Chia AJL, Randle LE, Hayes JD, Kitteringham NR, Park BK. The hepatotoxic metabolite of acetaminophen directly activates the Keap1-Nrf2 cell defense system. Hepatology 2008; 48:1292-301. [PMID: 18785192 DOI: 10.1002/hep.22472] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
UNLABELLED The transcription factor Nrf2 regulates the expression of numerous cytoprotective genes in mammalian cells. We have demonstrated previously that acetaminophen activates Nrf2 in mouse liver following administration of non-hepatotoxic and hepatotoxic doses in vivo, implying that Nrf2 may have an important role in the protection against drug-induced liver injury. Nrf2 activation has been proposed to occur through the modification of cysteine residues within Keap1, the cytosolic repressor of Nrf2. We hypothesized that acetaminophen activates Nrf2 via the formation of its reactive metabolite N-acetyl-p-benzoquinoneimine (NAPQI), which may disrupt the repression of Nrf2 through the modification of cysteine residues within Keap1. Here, we show that NAPQI can directly activate the Nrf2 pathway in mouse liver cells, inducing an adaptive defense response that is antagonized by RNA interference targeted against Nrf2. Furthermore, mass spectrometric analysis shows that NAPQI selectively modifies cysteine residues in Keap1, both in recombinant protein in vitro and in cells ectopically expressing Keap1. Using this cell-based model, we demonstrate that activation of Nrf2 by NAPQI and a panel of probe molecules [dexamethasone 21-mesylate, 15-deoxy-Delta-((12,14))-prostaglandin J(2), 2,4-dinitrochlorobenzene, and iodoacetamide] correlates with the selective modification of cysteine residues located within the intervening region of Keap1. However, substantial depletion of glutathione (to less than 15% of basal levels) by buthionine sulfoximine, which does not directly modify Keap1, is also sufficient to activate Nrf2. CONCLUSION Nrf2 can be activated via the direct modification of cysteine residues located within the intervening region of Keap1, but also via the substantial depletion of glutathione without the requirement for direct modification of Keap1. It is possible that both of these mechanisms contribute to the activation of Nrf2 by acetaminophen.
Collapse
Affiliation(s)
- Ian M Copple
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kuvandik G, Duru M, Nacar A, Yonden Z, Helvaci R, Koc A, Kozlu T, Kaya H, Sogüt S. Effects of Erdosteine on Acetaminophen-induced Hepatotoxicity in Rats. Toxicol Pathol 2008; 36:714-9. [DOI: 10.1177/0192623308320800] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated the effects of erdosteine on acetaminophen (APAP)-induced hepatotoxicity in rats. Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), AST (aspartate aminotransferase), and ALT (alanine transaminase) activities, and malonyldialdehyde (MDA) and nitric oxide levels as oxidant/antioxidant biochemical parameters were investigated with light microscopic evaluation in adult female Wistar Albino rats. APAP administration produced a decrease in hepatic SOD, CAT, and GSH-Px activities, and coadministration of erdosteine (150 and 300 mg/kg) resulted in increases in the activities. MDA and NO levels increased in the APAP group, and erdosteine treatments prevented these increases. Significant elevations in serum AST and ALT levels were observed in the APAP group, and when erdosteine and APAP were coadministered, their serum levels were close to those in the control group. Light microscopic evaluation of livers showed that there were remarkable centrilobular (zone III) hepatic necrosis and mild to moderate sinusoidal congestion in the APAP group, whereas in the erdosteine group, cellular necrosis was minimal and the hepatocytes maintained a better morphology when compared to the APAP group. Erdosteine prevented APAP-induced liver injury and toxic side effects probably through the antioxidant and radical scavenging effects of erdosteine.
Collapse
Affiliation(s)
| | - Mehmet Duru
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Ahmet Nacar
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Zafer Yonden
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Rami Helvaci
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Ahmet Koc
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Tolunay Kozlu
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Hasan Kaya
- Mustafa Kemal University Medical School, Hatay, Turkey
| | - Sadik Sogüt
- Mustafa Kemal University Medical School, Hatay, Turkey
| |
Collapse
|
44
|
Cali JJ, Niles A, Valley MP, O'Brien MA, Riss TL, Shultz J. Bioluminescent assays for ADMET. Expert Opin Drug Metab Toxicol 2008; 4:103-20. [PMID: 18370862 DOI: 10.1517/17425255.4.1.103] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Bioluminescent assays couple a limiting component of a luciferase-catalyzed photon-emitting reaction to a variable parameter of interest, while holding the other components constant or non-limiting. In this way light output varies with the parameter of interest. This review describes three bioluminescent assay types that use firefly luciferase to measure properties of drugs and other xenobiotics which affect their absorption, distribution, metabolism, elimination and toxicity. First, levels of the luciferase enzyme itself are measured in gene reporter assays that place a luciferase cDNA under the control of regulatory sequences from ADMET-related genes. This approach identifies activators of nuclear receptors that regulate expression of genes encoding drug-metabolizing enzymes and drug transporters. Second, drug effects on enzyme activities are monitored with luminogenic probe substrates that are inactive derivatives of the luciferase substrate luciferin. The enzymes of interest convert the substrates to free luciferin, which is detected in a second reaction with luciferase. This approach is used with the drug-metabolizing CYP and monoamine oxidase enzymes, apoptosis-associated caspase proteases, a marker protease for non-viable cells and with glutathione-S-transferase to measure glutathione levels in cell lysates. Third, ATP concentration is monitored as a marker of cell viability or cell death and as a way of identifying substrates for the ATP-dependent drug transporter, P-glycoprotein. Luciferase activity is measured in the presence of a sample that supplies the requisite luciferase substrate, ATP, so that light output varies with ATP concentration. The bioluminescent ADMET assays are rapid and sensitive, amenable to automated high-throughput applications and offer significant advantages over alternative methods.
Collapse
Affiliation(s)
- James J Cali
- Promega Corp., 2800 Woods Hollow Road, Madison, WI 53711, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
The innate immune system represents a critical first line of host response to infectious, injurious and inflammatory insults. NKT cells (natural killer T-cells) are an important, but relatively poorly understood, component of the innate immune response. Moreover, NKT cells are enriched within the liver, suggesting that within the hepatic compartment NKT cells probably fulfil important roles in the modulation of the immune response to infection or injury. NKT cells are characterized by their rapid activation and secretion of large amounts of numerous types of cytokines, including those within the Th1-type, Th2-type and Th17-type groups, which in turn can interact with a multitude of other cell types within the liver. In addition, NKT cells are capable of participating in a wide array of effector functions with regards to other cell types via NKT cell-surface-molecule expression [e.g. FASL (FAS ligand) and CD40L (CD40 ligand)] and the release of mediators (e.g. perforin and granzyme) contained in cellular granules, which in turn can activate or destroy other cells (i.e. immune or parenchymal cells) within the liver. Given the huge scope of potential actions that can be mediated by NKT cells, it has become increasingly apparent that NKT cells may fulfil both beneficial (e.g. clearance of virally infected cells) and harmful (e.g. induction of autoimmunity) roles in the setting of liver disease. This review will outline the possible roles which may be played by NKT cells in the setting of specific liver diseases or conditions, and will discuss the NKT cell in the context of its role as either a ‘friend’ or a ‘foe’ with respect to the outcome of these liver disorders.
Collapse
|
46
|
Whyte IM, Francis B, Dawson AH. Safety and efficacy of intravenous N-acetylcysteine for acetaminophen overdose: analysis of the Hunter Area Toxicology Service (HATS) database. Curr Med Res Opin 2007; 23:2359-68. [PMID: 17705945 DOI: 10.1185/030079907x219715] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Acetaminophen (N-acetyl-p-aminophenyl; APAP) is the leading drug used in self-poisoning and frequently causes hepatotoxicity, including acute liver failure. OBJECTIVE To provide descriptive data on the safety and efficacy of intravenous N-acetylcysteine (IV-NAC) in the treatment of APAP toxicity, based on information in the Hunter Area Toxicology Service (HATS) database involving residents of the Greater Newcastle Area of New South Wales, Australia. METHODS This was a retrospective analysis of all APAP overdoses from January 1987 to January 2003. Data were collected prospectively according to a published protocol and included patient characteristics, exposures to APAP and other potential toxins, treatments, and outcomes. Primary safety/tolerability endpoints included the mortality rate and incidence of adverse drug reactions, while efficacy endpoints included alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. RESULTS Of 1749 patients, 399 (22.8%) were treated with IV-NAC. Of these, 37 (9.3%) had an adverse drug reaction to IV-NAC, of which seven (1.8% of total) were anaphylactoid. There were five deaths in hospital (mortality rate = 0.3%), including two attributed to APAP (0.1%) and none to IV-NAC. Of 64 patients who were treated with IV-NAC within 8 hours after APAP ingestion and had available ALT/AST data, two (3.1%) developed hepatotoxicity (AST/ALT > 1000 IU/L) compared with 32 (25%) of 128 patients receiving IV-NAC > 8 hours after APAP ingestion (p = 0.0002). A total of 26 patients (15.6%) receiving IV-NAC treatment within 8 hours after APAP ingestion had hospitalization stays > 48 hours compared with 70 (33.3%) receiving IV-NAC > 8 hours after ingestion (p < 0.0001). CONCLUSIONS For patients with APAP overdose seen in the HATS database of New South Wales, Australia, in-hospital death was infrequent (< 1%) and hepatotoxicity was significantly less likely when IV-NAC was administered within 8 hours after APAP ingestion compared with longer intervals (p < 0.01). As a descriptive retrospective database analysis, this study could not exclude certain sources of bias, including temporal changes over the 16-year course of data collection in the use of IV-NAC and low ascertainment of mild, self-limiting reactions to IV-NAC.
Collapse
Affiliation(s)
- Ian M Whyte
- Department of Clinical Toxicology and Pharmacology, Newcastle Mater Misericordiae Hospital, Newcastle, Australia
| | | | | |
Collapse
|
47
|
Tang W. Drug metabolite profiling and elucidation of drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2007; 3:407-20. [PMID: 17539747 DOI: 10.1517/17425255.3.3.407] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Drug metabolism studies, together with pathologic and histologic evaluation, provide critical data sets to help understand mechanisms underlying drug-related hepatotoxicity. A common practice is to trace morphologic changes resulting from liver injury back to perturbation of biochemical processes and to identify drug metabolites that affect those processes as possible culprits. This strategy can be illustrated in efforts of elucidating the cause of acetaminophen-, troglitazone- and valproic acid-induced hepatic necrosis, microvesicular steatosis and cholestasis with the aid of information from qualitative and quantitative analysis of metabolites. From a pharmaceutical research perspective, metabolite profiling represents an important function because a structure-activity relationship is essential to rational drug design. In addition, drugs are known to induce idiosyncratic hepatotoxicity, which usually escapes the detection by preclinical safety assessment and clinical trials. This issue is addressed, at present, by eliminating those molecules that are prone to metabolic bioactivation, based on the concept that formation of electrophilic metabolites triggers covalent protein modification and subsequent organ toxicity. Although pragmatic, such an approach has its limitations as a linear correlation does not exist between toxicity and the extent of bioactivation. It may be possible in the future that the advance of proteomics, metabonomics and genomics would pave the way leading to personalized medication in which beneficial effect of a drug is maximized, whereas toxicity risk is minimized.
Collapse
Affiliation(s)
- Wei Tang
- Merck Research Laboratories, Department of Drug Metabolism, Rahway, NJ 07065-0900, USA.
| |
Collapse
|
48
|
Mitochondrial involvement in drug-induced hepatic injury. Chem Biol Interact 2006; 163:145-59. [DOI: 10.1016/j.cbi.2006.06.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 06/13/2006] [Accepted: 06/14/2006] [Indexed: 01/22/2023]
|
49
|
Rajaraman G, Chen J, Chang TKH. Ginkgolide A contributes to the potentiation of acetaminophen toxicity by Ginkgo biloba extract in primary cultures of rat hepatocytes. Toxicol Appl Pharmacol 2006; 217:225-33. [PMID: 17045319 DOI: 10.1016/j.taap.2006.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2006] [Revised: 09/01/2006] [Accepted: 09/05/2006] [Indexed: 11/18/2022]
Abstract
The present cell culture study investigated the effect of Ginkgo biloba extract pretreatment on acetaminophen toxicity and assessed the role of ginkgolide A and cytochrome P450 3A (CYP3A) in hepatocytes isolated from adult male Long-Evans rats provided ad libitum with a standard diet. Acetaminophen (7.5-25 mM for 24 h) conferred hepatocyte toxicity, as determined by the lactate dehydrogenase (LDH) assay. G. biloba extract alone increased LDH leakage in hepatocytes at concentrations > or =75 mug/ml and > or =750 mug/ml after a 72 h and 24 h treatment period, respectively. G. biloba extract (25 or 50 mug/ml once every 24 h for 72 h) potentiated LDH leakage by acetaminophen (10 mM for 24 h; added at 48 h after initiation of extract pretreatment). The effect was confirmed by a decrease in [(14)C]-leucine incorporation. At the level present in a modulating concentration (50 mug/ml) of the extract, ginkgolide A (0.55 mug/ml), which increased CYP3A23 mRNA levels and CYP3A-mediated enzyme activity, accounted for part but not all of the potentiating effect of the extract on acetaminophen toxicity. This occurred as a result of CYP3A induction by ginkgolide A because triacetyloleandomycin (TAO), a specific inhibitor of CYP3A catalytic activity, completely blocked the effect of ginkgolide A. Ginkgolide B, ginkgolide C, ginkgolide J, quercetin, kaempferol, isorhamnetin, and isorhamnetin-3-O-rutinoside did not alter the extent of LDH leakage by acetaminophen. In summary, G. biloba pretreatment potentiated acetaminophen toxicity in cultured rat hepatocytes and ginkgolide A contributed to this novel effect of the extract by inducing CYP3A.
Collapse
Affiliation(s)
- Ganesh Rajaraman
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | |
Collapse
|