1
|
El-Abassy OM, Fawzy MG, Kamel EB. Two chromatographic methods for analyzing paracetamol in spiked human plasma with its toxic metabolite, N-acetyl parabenzoquinone imine and its antidote, N-acetyl-L-cysteine. Sci Rep 2025; 15:4119. [PMID: 39900950 PMCID: PMC11791058 DOI: 10.1038/s41598-025-86070-3] [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: 08/24/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025] Open
Abstract
Acetaminophen, also known as paracetamol (APAP), is a highly utilized pharmaceutical agent on a global scale, particularly in the field of pediatrics. Regrettably, an overdose of APAP, resulting from the predominant oxidation, has the potential to trigger acute liver injury. The study's goal was to find an easy, accurate, and selective way to measure APAP, N-acetyl para benzoquinone imine (NAPQI) (an APAP metabolite that is harmful), and N-acetyl-L-cysteine (NAC) (an antidote). Two different chromatographic methods were used. The HPTLC method, which used silica gel 60 F254 as a stationary phase and a developing liquid made up of methanol, ethyl acetate, and glacial acetic acid (8:2:0.2, v/v/v) and a UV detection at 254 nm. The HPLC method was developed using a mobile phase consisting of water, methanol, and formic acid in a proportion of (70:30:0.15, v/v/v). The stationary phase used in the approach was a C18 column. Analytes quantification was established utilizing a UV detector operating at a wavelength of 254 nm. The present methods make it possible to measure the amount of APAP in plasma samples. When it comes to pharmacokinetics or medication levels in children's plasma, for example, this may be also very helpful. The current methods can quantify NAPQI, which is helpful in figuring out drug concentrations in individuals with APAP intoxication diagnoses. Additionally, the current approaches can estimate NAC as an antidote; as a result, this study is a complete study because it can analyse drug, toxic metabolite, and antidote in one analytical run. Using the innovative blue applicability grade index software, which measures the practicality of procedures, both methodologies were compared with a reported methods. Additionally, the achievement of the eco-friendliness profile of the designed procedures was assessed. Both techniques passed the ICH validation tests.
Collapse
Affiliation(s)
- Omar M El-Abassy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt.
| | - Michael Gamal Fawzy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| | - Ebraam B Kamel
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| |
Collapse
|
2
|
Rousar T, Handl J, Capek J, Nyvltova P, Rousarova E, Kubat M, Smid L, Vanova J, Malinak D, Musilek K, Cesla P. Cysteine conjugates of acetaminophen and p-aminophenol are potent inducers of cellular impairment in human proximal tubular kidney HK-2 cells. Arch Toxicol 2023; 97:2943-2954. [PMID: 37639014 PMCID: PMC10504157 DOI: 10.1007/s00204-023-03569-2] [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: 05/29/2023] [Accepted: 08/02/2023] [Indexed: 08/29/2023]
Abstract
Acetaminophen (APAP) belong among the most used analgesics and antipyretics. It is structurally derived from p-aminophenol (PAP), a potent inducer of kidney toxicity. Both compounds can be metabolized to oxidation products and conjugated with glutathione. The glutathione-conjugates can be cleaved to provide cysteine conjugates considered as generally nontoxic. The aim of the present report was to synthesize and to purify both APAP- and PAP-cysteine conjugates and, as the first study at all, to evaluate their biological effects in human kidney HK-2 cells in comparison to parent compounds. HK-2 cells were treated with tested compounds (0-1000 µM) for up to 24 h. Cell viability, glutathione levels, ROS production and mitochondrial function were determined. After 24 h, we found that both APAP- and PAP-cysteine conjugates (1 mM) were capable to induce harmful cellular damage observed as a decrease of glutathione levels to 10% and 0%, respectively, compared to control cells. In addition, we detected the disappearance of mitochondrial membrane potential in these cells. In the case of PAP-cysteine, the extent of cellular impairment was comparable to that induced by PAP at similar doses. On the other hand, 1 mM APAP-cysteine induced even larger damage of HK-2 cells compared to 1 mM APAP after 6 or 24 h. We conclude that cysteine conjugates with aminophenol are potent inducers of oxidative stress causing significant injury in kidney cells. Thus, the harmful effects cysteine-aminophenolic conjugates ought to be considered in the description of APAP or PAP toxicity.
Collapse
Affiliation(s)
- Tomas Rousar
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic.
| | - Jiri Handl
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Jan Capek
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Pavlina Nyvltova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Erika Rousarova
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Miroslav Kubat
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Lenka Smid
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - Jana Vanova
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| | - David Malinak
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Petr Cesla
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentska 95, 532 10, Pardubice, Czech Republic
| |
Collapse
|
3
|
Mally A, Jarzina S. Mapping Adverse Outcome Pathways for Kidney Injury as a Basis for the Development of Mechanism-Based Animal-Sparing Approaches to Assessment of Nephrotoxicity. FRONTIERS IN TOXICOLOGY 2022; 4:863643. [PMID: 35785263 PMCID: PMC9242087 DOI: 10.3389/ftox.2022.863643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
In line with recent OECD activities on the use of AOPs in developing Integrated Approaches to Testing and Assessment (IATAs), it is expected that systematic mapping of AOPs leading to systemic toxicity may provide a mechanistic framework for the development and implementation of mechanism-based in vitro endpoints. These may form part of an integrated testing strategy to reduce the need for repeated dose toxicity studies. Focusing on kidney and in particular the proximal tubule epithelium as a key target site of chemical-induced injury, the overall aim of this work is to contribute to building a network of AOPs leading to nephrotoxicity. Current mechanistic understanding of kidney injury initiated by 1) inhibition of mitochondrial DNA polymerase γ (mtDNA Polγ), 2) receptor mediated endocytosis and lysosomal overload, and 3) covalent protein binding, which all present fairly well established, common mechanisms by which certain chemicals or drugs may cause nephrotoxicity, is presented and systematically captured in a formal description of AOPs in line with the OECD AOP development programme and in accordance with the harmonized terminology provided by the Collaborative Adverse Outcome Pathway Wiki. The relative level of confidence in the established AOPs is assessed based on evolved Bradford-Hill weight of evidence considerations of biological plausibility, essentiality and empirical support (temporal and dose-response concordance).
Collapse
|
4
|
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]
|
5
|
Un H, Ugan RA, Gurbuz MA, Bayir Y, Kahramanlar A, Kaya G, Cadirci E, Halici Z. Phloretin and phloridzin guard against cisplatin-induced nephrotoxicity in mice through inhibiting oxidative stress and inflammation. Life Sci 2020; 266:118869. [PMID: 33309722 DOI: 10.1016/j.lfs.2020.118869] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
AIM Cisplatin (Cis) is widely used chemotherapeutic and has some serious side effects as nephrotoxicity. Phloretin (PH) and Phloridzin (PZ) are known their anti-oxidant anti-inflammatory effects. We aimed to examine the protective effects of PH and PZ on cisplatin-induced nephrotoxicity. MAIN METHODS Totally, 48 Balb/C female mice were separated into eight groups (n = 6). First day, single dose of cisplatin (20 mg/kg intraperitoneal) was administered to induce toxicity. PH and PZ were given (50 and 100 mg/kg orally) to treatment groups during 3 days. After the experimental procedures serum renal function enzymes (BUN and Creatinine), oxidative parameters (SOD, GSH and MDA), nuclear agent NFKβ, inflammatory cytokines (Tnf-α and IL1β) and HSP70 expressions and histopathological assessments were analyzed. KEY FINDINGS Serum enzymes, tissue cytokines and oxidative stress were increased after the Cis treatment. PH and PZ treatments normalized all parameters compared to Cis administrated group. After the treatments, SOD activities and GSH levels were increased while MDA levels were decreased. PH and PZ treatments decreased Tnf-α, IL1β and NFKβ mRNA expressions. Cis significantly increased the HSP70 expression while PH and PZ administrations significantly decreased. Similar the biochemical and molecular results, PH and PZ showed positive effects on tissue pathological parameters. Cisplatin cause a lot of abnormal structures as tubular and glomeruli damages on the kidney. SIGNIFICANCE PH and PZ play important physiological roles in the prevention of nephrotoxicity. Antioxidant and anti-inflammatory effects of PH and PZ demonstrated visible protective effects in the cisplatin-induced nephrotoxicity model.
Collapse
Affiliation(s)
- Harun Un
- Agri Ibrahim Cecen University, Faculty of Pharmacy, Department of Biochemistry, Agri, Turkey.
| | - Rustem Anil Ugan
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, Erzurum, Turkey
| | - Muhammet Ali Gurbuz
- Ataturk University, Faculty of Medicine, Department of Histology and Embryology, Erzurum, Turkey
| | - Yasin Bayir
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Aysenur Kahramanlar
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Gokce Kaya
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
6
|
Baillie TA. Approaches to mitigate the risk of serious adverse reactions in covalent drug design. Expert Opin Drug Discov 2020; 16:275-287. [PMID: 33006907 DOI: 10.1080/17460441.2021.1832079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Covalent inhibition of target proteins using high affinity ligands bearing weakly electrophilic warheads is being adopted increasingly as design strategy in the discovery of novel therapeutics, and several covalent drugs have now received regulatory approval for indications in oncology. Experience to date with targeted covalent inhibitors has led to a number of design principles that underlie the safety and efficacy of this increasingly important class of molecules. AREAS COVERED A review is provided of the current status of the covalent drug approach, emphasizing the unique benefits and attendant risks associated with reversible and irreversible binders. Areas of application beyond inhibition of tyrosine kinases are presented, and design considerations to de-risk covalent inhibitors with respect to undesirable off-target effects are discussed. EXPERT OPINION High selectivity for the intended protein target has emerged as a key consideration in mitigating safety risks associated with widespread proteome reactivity. Powerful chemical proteomics-based techniques are now available to assess selectivity in a drug discovery setting. Optimizing pharmacokinetics to capitalize on the intrinsically high potency of covalent drugs should lead to low daily doses and greater safety margins, while minimizing susceptibility to metabolic activation likewise will attenuate the risk of covalent drug toxicity.
Collapse
Affiliation(s)
- Thomas A Baillie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington Seattle, Seattle, WA, USA
| |
Collapse
|
7
|
Abstract
Many potentially toxic electrophilic xenobiotics and some endogenous compounds are detoxified by conversion to the corresponding glutathione S-conjugate, which is metabolized to the N-acetylcysteine S-conjugate (mercapturate) and excreted. Some mercapturate pathway components, however, are toxic. Bioactivation (toxification) may occur when the glutathione S-conjugate (or mercapturate) is converted to a cysteine S-conjugate that undergoes a β-lyase reaction. If the sulfhydryl-containing fragment produced in this reaction is reactive, toxicity may ensue. Some drugs and halogenated workplace/environmental contaminants are bioactivated by this mechanism. On the other hand, cysteine S-conjugate β-lyases occur in nature as a means of generating some biologically useful sulfhydryl-containing compounds.
Collapse
|
8
|
Kobayashi S, Lee J, Takao T, Fujii J. Increased ophthalmic acid production is supported by amino acid catabolism under fasting conditions in mice. Biochem Biophys Res Commun 2017; 491:649-655. [DOI: 10.1016/j.bbrc.2017.07.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 01/18/2023]
|
9
|
Rankin GO, Tyree C, Pope D, Tate J, Racine C, Anestis DK, Brown KC, Dial M, Valentovic MA. Role of Free Radicals and Biotransformation in Trichloronitrobenzene-Induced Nephrotoxicity In Vitro. Int J Mol Sci 2017; 18:ijms18061165. [PMID: 28561793 PMCID: PMC5485989 DOI: 10.3390/ijms18061165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/12/2017] [Accepted: 05/24/2017] [Indexed: 11/25/2022] Open
Abstract
This study determined the comparative nephrotoxic potential of four trichloronitrobenzenes (TCNBs) (2,3,4-; 2,4,5-; 2,4,6-; and 3,4,5-TCNB) and explored the effects of antioxidants and biotransformation inhibitors on TCNB-induced cytotoxicity in isolated renal cortical cells (IRCC) from male Fischer 344 rats. IRCC were incubated with a TCNB up to 1.0 mM for 15–120 min. Pretreatment with an antioxidant or cytochrome P450 (CYP), flavin monooxygenase (FMO), or peroxidase inhibitor was used in some experiments. Among the four TCNBs, the order of decreasing nephrotoxic potential was approximately 3,4,5- > 2,4,6- > 2,3,4- > 2,4,5-TCNB. The four TCNBs exhibited a similar profile of attenuation of cytotoxicity in response to antioxidant pretreatments. 2,3,4- and 3,4,5-TCNB cytotoxicity was attenuated by most of the biotransformation inhibitors tested, 2,4,5-TCNB cytotoxicity was only inhibited by isoniazid (CYP 2E1 inhibitor), and 2,4,6-TCNB-induced cytotoxicity was inhibited by one CYP inhibitor, one FMO inhibitor, and one peroxidase inhibitor. All of the CYP specific inhibitors tested offered some attenuation of 3,4,5-TCNB cytotoxicity. These results indicate that 3,4,5-TCNB is the most potent nephrotoxicant, free radicals play a role in the TCNB cytotoxicity, and the role of biotransformation in TCNB nephrotoxicity in vitro is variable and dependent on the position of the chloro groups.
Collapse
Affiliation(s)
- Gary O Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Connor Tyree
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Deborah Pope
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Jordan Tate
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Christopher Racine
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Dianne K Anestis
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Kathleen C Brown
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Mason Dial
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Monica A Valentovic
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
10
|
Gan J, Ma S, Zhang D. Non-cytochrome P450-mediated bioactivation and its toxicological relevance. Drug Metab Rev 2016; 48:473-501. [DOI: 10.1080/03602532.2016.1225756] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
11
|
Fan PW, Zhang D, Halladay JS, Driscoll JP, Khojasteh SC. Going Beyond Common Drug Metabolizing Enzymes: Case Studies of Biotransformation Involving Aldehyde Oxidase, γ-Glutamyl Transpeptidase, Cathepsin B, Flavin-Containing Monooxygenase, and ADP-Ribosyltransferase. Drug Metab Dispos 2016; 44:1253-61. [PMID: 27117704 DOI: 10.1124/dmd.116.070169] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/25/2016] [Indexed: 11/22/2022] Open
Abstract
The significant roles that cytochrome P450 (P450) and UDP-glucuronosyl transferase (UGT) enzymes play in drug discovery cannot be ignored, and these enzyme systems are commonly examined during drug optimization using liver microsomes or hepatocytes. At the same time, other drug-metabolizing enzymes have a role in the metabolism of drugs and can lead to challenges in drug optimization that could be mitigated if the contributions of these enzymes were better understood. We present examples (mostly from Genentech) of five different non-P450 and non-UGT enzymes that contribute to the metabolic clearance or bioactivation of drugs and drug candidates. Aldehyde oxidase mediates a unique amide hydrolysis of GDC-0834 (N-[3-[6-[4-[(2R)-1,4-dimethyl-3-oxopiperazin-2-yl]anilino]-4-methyl-5-oxopyrazin-2-yl]-2-methylphenyl]-4,5,6,7-tetrahydro-1-benzothiophene-2-carboxamide), leading to high clearance of the drug. Likewise, the rodent-specific ribose conjugation by ADP-ribosyltransferase leads to high clearance of an interleukin-2-inducible T-cell kinase inhibitor. Metabolic reactions by flavin-containing monooxygenases (FMO) are easily mistaken for P450-mediated metabolism such as oxidative defluorination of 4-fluoro-N-methylaniline by FMO. Gamma-glutamyl transpeptidase is involved in the initial hydrolysis of glutathione metabolites, leading to formation of proximate toxins and nephrotoxicity, as is observed with cisplatin in the clinic, or renal toxicity, as is observed with efavirenz in rodents. Finally, cathepsin B is a lysosomal enzyme that is highly expressed in human tumors and has been targeted to release potent cytotoxins, as in the case of brentuximab vedotin. These examples of non-P450- and non-UGT-mediated metabolism show that a more complete understanding of drug metabolizing enzymes allows for better insight into the fate of drugs and improved design strategies of molecules in drug discovery.
Collapse
Affiliation(s)
- Peter W Fan
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco (P.W.F., D.Z., S.C.K.); Anacor Pharmaceuticals, Inc., Palo Alto (J.S.H.); MyoKardia, Inc., South San Francisco (J.P.D.), California
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco (P.W.F., D.Z., S.C.K.); Anacor Pharmaceuticals, Inc., Palo Alto (J.S.H.); MyoKardia, Inc., South San Francisco (J.P.D.), California
| | - Jason S Halladay
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco (P.W.F., D.Z., S.C.K.); Anacor Pharmaceuticals, Inc., Palo Alto (J.S.H.); MyoKardia, Inc., South San Francisco (J.P.D.), California
| | - James P Driscoll
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco (P.W.F., D.Z., S.C.K.); Anacor Pharmaceuticals, Inc., Palo Alto (J.S.H.); MyoKardia, Inc., South San Francisco (J.P.D.), California
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco (P.W.F., D.Z., S.C.K.); Anacor Pharmaceuticals, Inc., Palo Alto (J.S.H.); MyoKardia, Inc., South San Francisco (J.P.D.), California
| |
Collapse
|
12
|
Guo X, Chen L, Chen J, Xie P, Li S, He J, Li W, Fan H, Yu D, Zeng C. Quantitatively evaluating detoxification of the hepatotoxic microcystin-LR through the glutathione (GSH) pathway in SD rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:19273-19284. [PMID: 26490924 DOI: 10.1007/s11356-015-5531-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/05/2015] [Indexed: 06/05/2023]
Abstract
Glutathione (GSH) plays crucial roles in antioxidant defense and detoxification metabolism of microcystin-LR (MC-LR). However, the detoxification process of MC-LR in mammals remains largely unknown. This paper, for the first time, quantitatively analyzes MC-LR and its GSH pathway metabolites (MC-LR-GSH and MC-LR-Cys) in the liver of Sprague-Dawley (SD) rat after MC-LR exposure. Rats received intraperitoneal (i.p.) injection of 0.25 and 0.5 lethal dose 50 (LD50) of MC-LR with or without pretreatment of buthionine-(S,R)-sulfoximine (BSO), an inhibitor of GSH synthesis. The contents of MC-LR-GSH were relatively low during the experiment; however, the ratio of MC-LR-Cys to MC-LR reached as high as 6.65 in 0.5 LD50 group. These results demonstrated that MC-LR-GSH could be converted to MC-LR-Cys efficiently, and this metabolic rule was in agreement with the data of aquatic animals previously reported. MC-LR contents were much higher in BSO + MC-LR-treated groups than in the single MC-LR-treated groups. Moreover, the ratio of MC-LR-Cys to MC-LR decreased significantly after BSO pretreatment, suggesting that the depletion of GSH induced by BSO reduced the detoxification of MCs. Moreover, MC-LR remarkably induced liver damage, and the effects were more pronounced in BSO pretreatment groups. In conclusion, this study verifies the role of GSH in the detoxification of MC-LR and furthers our understanding of the biochemical mechanism for SD rats to counteract toxic cyanobacteria.
Collapse
Affiliation(s)
- Xiaochun Guo
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
| | - Liang Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China.
| | - Ping Xie
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China.
| | - Shangchun Li
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun He
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
| | - Wei Li
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huihui Fan
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
| | - Dezhao Yu
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cheng Zeng
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, China
| |
Collapse
|
13
|
Three common pathways of nephrotoxicity induced by halogenated alkenes. Cell Biol Toxicol 2015; 31:1-13. [DOI: 10.1007/s10565-015-9293-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/29/2015] [Indexed: 12/13/2022]
|
14
|
Wu L, Wang Q, Tao M, Chen J, Ma Z, Xie P. Preliminary study of the distribution and accumulation of GSH/Cys metabolites of hepatotoxic microcystins-RR in common carp from a lake with protracted cyanobacterial bloom (Lake Taihu, China). BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2013; 90:382-386. [PMID: 23272327 DOI: 10.1007/s00128-012-0947-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 12/19/2012] [Indexed: 06/01/2023]
Abstract
Tissue distributions and seasonal dynamics of glutathione and cysteine S-conjugates of microcystin-RR in feral fish from Lake Taihu were studied. High MC-RR-Cys was found in tissues, Mean MC-RR-Cys in kidney (0.253 μg g(-1) DW) was 4 times of that in liver (0.063 μg g(-1) DW). Ratios of MC-RR-Cys/MC-RR in liver/kidney reached as high as 5.3/39.8, respectively, meanwhile, kidney showed low accumulation of MC-RR and higher formation efficiency of MC-RR-Cys than liver (7.51× to liver), this suggested that MC-RR-Cys were significantly accumulated with the depletion of MC-RR, and it was selectively biotransformed to MC-RR-Cys in kidney for further excretion.
Collapse
Affiliation(s)
- Laiyan Wu
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7th, Wuhan, 430072, People's Republic of China
| | | | | | | | | | | |
Collapse
|
15
|
Hausheer F, Ding D, Shanmugarajah D, Leverett B, Huang Q, Chen X, Kochat H, Ayala P, Petluru P, Parker A. Accumulation of BNP7787 in Human Renal Proximal Tubule Cells. J Pharm Sci 2011; 100:3977-84. [DOI: 10.1002/jps.22510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 01/19/2011] [Accepted: 01/19/2011] [Indexed: 11/09/2022]
|
16
|
Sevgiler Y, Üner N. Tissue-specific effects of fenthion on glutathione metabolism modulated by NAC and BSO inOreochromis niloticus. Drug Chem Toxicol 2010; 33:348-56. [DOI: 10.3109/01480541003734048] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Mechanistic study of BNP7787-mediated cisplatin nephroprotection: modulation of human aminopeptidase N. Cancer Chemother Pharmacol 2010; 67:381-91. [PMID: 20440617 DOI: 10.1007/s00280-010-1333-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE Previous studies from our laboratory have identified a role for gamma-glutamyl transpeptidase (GGT) in BNP7787 (disodium 2,2'-dithio-bis ethane sulfonate, dimesna, Tavocept™)-mediated cisplatin nephroprotection. Dekant has proposed that gamma-glutamyl transpeptidase (GGT), aminopeptidase N (APN) and cysteine-conjugate-β-lyase (CCBL) comprise a multi-enzyme pathway that acts on xenobiotic-glutathione conjugates converting them to nephrotoxic metabolites. We report modulation of APN activity within this pathway by BNP7787-derived mesna-disulfide heteroconjugates. METHODS A fluorimetric assay was used to determine the effect of BNP7787, BNP7787-derived mesna-disulfide heteroconjugates, and the BNP7787 metabolite, mesna (sodium 2-mercaptoethane sulfonate), on the initial velocity and overall progress curve of the human APN reaction in vitro. RESULTS Neither BNP7787 nor mesna-cysteinyl-glutamate inhibited human APN. Select BNP7787-derived mesna-disulfide heteroconjugates (mesna-cysteine, mesna-glutathione, mesna-cysteinyl-glycine) and high concentrations of mesna inhibited APN activity. Allosteric effects on the enzyme progress curve outside of the linear initial velocity region were observed for mesna-cysteinyl-glycine, mesna-glutathione and mesna-cysteinyl-glutamate and appeared to be a function of having both mesna and di- or tri-peptide functionalities in one molecule. In situ-generated mesna-cisplatin conjugates were not a substrate for human APN. CONCLUSIONS BNP7787-mediated prevention or mitigation of cisplatin-induced nephrotoxicity may involve APN inhibition by certain BNP7787-derived mesna-disulfide heteroconjugates and appears correlated to the presence of a glycinate moiety and/or an anionic group. Two general mechanisms for BNP7787-mediated nephroprotection of cisplatin-induced nephrotoxicity involving the GGT, APN and CCBL nephrotoxigenic pathway are proposed which acting in a concerted and/or synergistic manner, and thereby prevent or mitigate cisplatin-induced renal toxicity.
Collapse
|
18
|
Veiga-da-Cunha M, Tyteca D, Stroobant V, Courtoy PJ, Opperdoes FR, Van Schaftingen E. Molecular identification of NAT8 as the enzyme that acetylates cysteine S-conjugates to mercapturic acids. J Biol Chem 2010; 285:18888-98. [PMID: 20392701 DOI: 10.1074/jbc.m110.110924] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Our goal was to identify the reaction catalyzed by NAT8 (N-acetyltransferase 8), a putative N-acetyltransferase homologous to the enzyme (NAT8L) that produces N-acetylaspartate in brain. The almost exclusive expression of NAT8 in kidney and liver and its predicted association with the endoplasmic reticulum suggested that it was cysteinyl-S-conjugate N-acetyltransferase, the microsomal enzyme that catalyzes the last step of mercapturic acid formation. In agreement, HEK293T extracts of cells overexpressing NAT8 catalyzed the N-acetylation of S-benzyl-L-cysteine and leukotriene E(4), two cysteine conjugates, but were inactive on other physiological amines or amino acids. Confocal microscopy indicated that NAT8 was associated with the endoplasmic reticulum. Neither of the two frequent single nucleotide polymorphisms found in NAT8, E104K nor F143S, changed the enzymatic activity or the expression of the protein by >or=2-fold, whereas a mutation (R149K) replacing an extremely conserved arginine suppressed the activity. Sequencing of genomic DNA and EST clones corresponding to the NAT8B gene, which resulted from duplication of the NAT8 gene in the primate lineage, disclosed the systematic presence of a premature stop codon at codon 16. Furthermore, truncated NAT8B and NAT8 proteins starting from the following methionine (Met-25) showed no cysteinyl-S-conjugate N-acetyltransferase activity when transfected in HEK293T cells. Taken together, these findings indicate that NAT8 is involved in mercapturic acid formation and confirm that NAT8B is an inactive gene in humans. NAT8 homologues are found in all vertebrate genomes, where they are often encoded by multiple, tandemly repeated genes as many other genes encoding xenobiotic metabolism enzymes.
Collapse
Affiliation(s)
- Maria Veiga-da-Cunha
- Laboratory of Physiological Chemistry, de Duve Institute and Université Catholique de Louvain, Avenue Hippocrate 75, B-1200 Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Many potentially toxic electrophiles react with glutathione to form glutathione S-conjugates in reactions catalyzed or enhanced by glutathione S-transferases. The glutathione S-conjugate is sequentially converted to the cysteinylglycine-, cysteine- and N-acetyl-cysteine S-conjugate (mercapturate). The mercapturate is generally more polar and water soluble than the parent electrophile and is readily excreted. Excretion of the mercapturate represents a detoxication mechanism. Some endogenous compounds, such as leukotrienes, prostaglandin (PG) A2, 15-deoxy-Δ12,14-PGJ2, and hydroxynonenal can also be metabolized to mercapturates and excreted. On occasion, however, formation of glutathione S- and cysteine S-conjugates are bioactivation events as the metabolites are mutagenic and/or cytotoxic. When the cysteine S-conjugate contains a strong electron-withdrawing group attached at the sulfur, it may be converted by cysteine S-conjugate β-lyases to pyruvate, ammonium and the original electrophile modified to contain an –SH group. If this modified electrophile is highly reactive then the enzymes of the mercapturate pathway together with the cysteine S-conjugate β-lyases constitute a bioactivation pathway. Some endogenous halogenated environmental contaminants and drugs are bioactivated by this mechanism. Recent studies suggest that coupling of enzymes of the mercapturate pathway to cysteine S-conjugate β-lyases may be more common in nature and more widespread in the metabolism of electrophilic xenobiotics than previously realized.
Collapse
|
20
|
Hausheer FH, Shanmugarajah D, Leverett BD, Chen X, Huang Q, Kochat H, Petluru PN, Parker AR. Mechanistic study of BNP7787-mediated cisplatin nephroprotection: modulation of gamma-glutamyl transpeptidase. Cancer Chemother Pharmacol 2009; 65:941-51. [PMID: 19714332 DOI: 10.1007/s00280-009-1101-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 07/29/2009] [Indexed: 11/28/2022]
Abstract
PURPOSE The mechanisms for cisplatin-induced renal cell injury have been the focus of intense investigation for many years with a view to provide a more effective and convenient form of nephroprotection. BNP7787 (disodium 2,2'-dithio-bis ethane sulfonate; dimesna, Tavocept), is a water-soluble disulfide investigational new drug that is undergoing clinical development for the prevention and mitigation of clinically important chemotherapy-induced toxicities associated with platinum-type chemotherapeutic agents. We hypothesized that part of BNP7787's mechanism of action (MOA) pertaining to the potential prevention of cisplatin-induced nephrotoxicity involves the inhibition of gamma-glutamyl transpeptidase (GGT) activity, mediated by BNP7787-derived mesna-disulfide heteroconjugates that contain a terminal gamma-glutamate moiety [e.g., mesna-glutathione (MSSGlutathione) and mesna-cysteinyl-glutamate (MSSCE)]. METHODS Inhibition studies were conducted on human and porcine GGT to determine the effect of mesna-disulfide heteroconjugates on the enzyme's activity in vitro. These studies utilized a fluorimetric assay that monitored the hydrolysis of L-gamma-glutamyl-7-amino-4-trifluoromethylcoumarin (GG-AFC) to AFC. RESULTS Mesna-disulfide heteroconjugates that contained gamma-glutamyl moieties were potent inhibitors of human and porcine GGT. An in situ-generated mesna-cisplatin conjugate was not a substrate for GGT. CONCLUSIONS The GGT xenobiotic metabolism pathway is postulated to be a major toxification pathway for cisplatin nephrotoxicity, and BNP7787 may play a novel and critical therapeutic role in the modulation of GGT activity. We further postulate that there are two general mechanisms for BNP7787-mediated nephroprotection against cisplatin-induced nephrotoxicity involving this pathway. First, the active BNP7787 pharmacophore, mesna, produces an inactive mesna-cisplatin conjugate that is not a substrate for the GGT toxification pathway (GGT xenobiotic metabolism pathway) and, second, BNP7787-derived mesna-disulfide heteroconjugates may serve as selective, potent inhibitors of GGT, possibly resulting in nephroprotection by a novel means.
Collapse
Affiliation(s)
- Frederick H Hausheer
- BioNumerik Pharmaceuticals, Inc., 8122 Datapoint Drive, Suite 1250, San Antonio, TX 78229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Moore LE, Wilson RT, Campleman SL. Lifestyle Factors, Exposures, Genetic Susceptibility, and Renal Cell Cancer Risk: A Review. Cancer Invest 2009; 23:240-55. [PMID: 15945510 DOI: 10.1081/cnv-200055962] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Malignant kidney tumors account for approximately 2% of all new primary cancer cases diagnosed in the United States, with an estimated 30,000 cases occurring annually. Although a variety of agents, chemical and biological, have been implicated as causal agents in the development of renal cell carcinoma (RCC), the etiology remains enigmatic. The strongest association has been developed between cigarette smoking and renal cancer however consistent, positive associations between RCC and obesity, diabetes, and hypertension have also been reported. In addition, more recent investigations of familial kidney cancer syndromes indicate that a strong genetic component contributes to RCC development. Several genes have been identified through investigation of familial kidney cancer syndromes. This review article describes recent trends in RCC incidence and the currently identifiable etiological causes that account for approximately half of the RCC cases diagnoses. The remainder of this review then focuses on additional risk factors that have thus far not been well examined but may be helpful in explaining the increasing incidence trends and the geographic or racial variation observed nationally and worldwide.
Collapse
Affiliation(s)
- Lee E Moore
- Occupational Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
22
|
Eaton DL, Daroff RB, Autrup H, Bridges J, Buffler P, Costa LG, Coyle J, McKhann G, Mobley WC, Nadel L, Neubert D, Schulte-Hermann R, Spencer PS. Review of the Toxicology of Chlorpyrifos With an Emphasis on Human Exposure and Neurodevelopment. Crit Rev Toxicol 2008; 38 Suppl 2:1-125. [PMID: 18726789 DOI: 10.1080/10408440802272158] [Citation(s) in RCA: 438] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Meijerman I, Beijnen JH, Schellens JH. Combined action and regulation of phase II enzymes and multidrug resistance proteins in multidrug resistance in cancer. Cancer Treat Rev 2008; 34:505-20. [DOI: 10.1016/j.ctrv.2008.03.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 02/11/2008] [Accepted: 03/01/2008] [Indexed: 01/16/2023]
|
24
|
Mechanism of chloroform-induced renal toxicity: non-involvement of hepatic cytochrome P450-dependent metabolism. Toxicol Appl Pharmacol 2007; 227:48-55. [PMID: 18031782 DOI: 10.1016/j.taap.2007.10.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 10/12/2007] [Accepted: 10/16/2007] [Indexed: 11/21/2022]
Abstract
Chloroform causes hepatic and renal toxicity in a number of species. In vitro studies have indicated that chloroform can be metabolized by P450 enzymes in the kidney to nephrotoxic intermediate, although direct in vivo evidence for the role of renal P450 in the nephrotoxicity has not been reported. This study was to determine whether chloroform renal toxicity persists in a mouse model with a liver-specific deletion of the P450 reductase (Cpr) gene (liver-Cpr-null). Chloroform-induced renal toxicity and chloroform tissue levels were compared between the liver-Cpr-null and wild-type mice at 24 h following differing doses of chloroform. At a chloroform dose of 150 mg/kg, the levels of blood urea nitrogen (BUN) were five times higher in the exposed group than in the vehicle-treated one for the liver-Cpr-null mice, but they were only slightly higher in the exposed group than in the vehicle-treated group for the wild-type mice. Severe lesions were found in the kidney of the liver-Cpr-null mice, while only mild lesions were found in the wild-type mice. At a chloroform dose of 300 mg/kg, severe kidney lesions were observed in both strains, yet the BUN levels were still higher in the liver-Cpr-null than in the wild-type mice. Higher chloroform levels were found in the tissues of the liver-Cpr-null mice. These findings indicated that loss of hepatic P450-dependent chloroform metabolism does not protect against chloroform-induced renal toxicity, suggesting that renal P450 enzymes play an essential role in chloroform renal toxicity.
Collapse
|
25
|
Bicker W, Lämmerhofer M, Genser D, Kiss H, Lindner W. A case study of acute human chlorpyrifos poisoning: Novel aspects on metabolism and toxicokinetics derived from liquid chromatography–tandem mass spectrometry analysis of urine samples. Toxicol Lett 2005; 159:235-51. [PMID: 15996837 DOI: 10.1016/j.toxlet.2005.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 05/23/2005] [Accepted: 05/25/2005] [Indexed: 01/19/2023]
Abstract
The metabolic fate of the organophosphorothioate-type insecticide chlorpyrifos (CP) in an acutely intoxicated 59 years old female was investigated by liquid chromatography-electrospray ionisation-tandem mass spectrometry (LC-ESI-MS/MS) analysis of urine samples. Fifteen metabolites of CP and its bioactivated intermediate chlorpyrifos-oxon (CPO), respectively, of which only three have been described in man so far, were identified on the basis of characteristic MS/MS transitions, precursor/product ion and/or neutral loss scans, chlorine isotopomer patterns, and partly by synthesis of reference compounds and subsequent structure confirmation. Three distinct biotransformation routes of CP are proposed: (1) cleavage reactions at the aromatic phosphoester bond, (2) cleavage reactions at the alkyl phosphoester bonds, and (3) glutathione (GSH) dependent nucleophilic substitution of the 6-chlorine at the aromatic moiety. Route (2) has not been reported in humans before and (3) is a hitherto completely unknown scheme of CP metabolism. Urinary markers of the latter were chiefly cysteine S-conjugates of mono-dechlorinated CP, CPO, mono-O-deethyl CP, and mono-O-deethyl CPO as well as the 6-mercapturic acid conjugate of 3,5-dichloro-2-pyridinol. The presence of 3,5-dichloro-6-methylthio-2-pyridinol as well as its O-glucuronide suggests further a cysteine S-conjugate beta-lyase mediated degradation. In addition to the qualitative LC-MS/MS screening the renal elimination profiles of the primary products of scheme (1), i.e. diethyl thiophosphate (DETP), diethyl phosphate (DEP), and 3,5,6-trichloro-2-pyridinol (TCP), were monitored over 14 days (n=21). A biphasic first-order excretion mechanism with half-lives of 21.5h (initial fast excretion phase) and 119.5h (terminal phase) for the sum of free DETP and DEP was found. TCP was hardly eliminated in its free form (O-glucuronide identified as phase II conjugate) and half-lives calculated for the total amount of TCP (acidic hydrolysis of urine samples) were 40.8 and 150.7h. The present study gives a more detailed view on the biotransformation of CP and together with the obtained kinetic data adds novel aspects to the limited knowledge of human metabolism of this xenobiotic, in particular at high dosage.
Collapse
Affiliation(s)
- Wolfgang Bicker
- Christian Doppler Laboratory for Molecular Recognition Materials, Department of Analytical and Food Chemistry, University of Vienna, Währinger Strasse 38, A-1090 Vienna, Austria
| | | | | | | | | |
Collapse
|
26
|
Jao SC, Chen J, Yang K, Li WS. Design of potent inhibitors for Schistosoma japonica glutathione S-transferase. Bioorg Med Chem 2005; 14:304-18. [PMID: 16275109 DOI: 10.1016/j.bmc.2005.07.077] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Revised: 07/22/2005] [Accepted: 07/22/2005] [Indexed: 11/18/2022]
Abstract
We implemented both structure-based drug design and the concept of polyvalency to discover a series of potent and unsymmetrical Schistosoma japonicum glutathione S-transferase (SjGST) inhibitors 10-12. This strategy achieved not only an excellent enhancement (10- to 490-fold) in the inhibitory potency, compared to the monofunctional analogues 1-5, but was also an effective modification by selecting a hydrophobic moiety with a flexible linker. The designed compounds with a low micromolar hit demonstrate special values in refining the new generation of SjGST inhibitors. The stoichiometry of the binding is one inhibitor molecule per SjGST monomer via isothermal titration calorimetric measurement.
Collapse
Affiliation(s)
- Shu-Chuan Jao
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | |
Collapse
|
27
|
Zhou S, Chan E, Duan W, Huang M, Chen YZ. Drug bioactivation, covalent binding to target proteins and toxicity relevance. Drug Metab Rev 2005; 37:41-213. [PMID: 15747500 DOI: 10.1081/dmr-200028812] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A number of therapeutic drugs with different structures and mechanisms of action have been reported to undergo metabolic activation by Phase I or Phase II drug-metabolizing enzymes. The bioactivation gives rise to reactive metabolites/intermediates, which readily confer covalent binding to various target proteins by nucleophilic substitution and/or Schiff's base mechanism. These drugs include analgesics (e.g., acetaminophen), antibacterial agents (e.g., sulfonamides and macrolide antibiotics), anticancer drugs (e.g., irinotecan), antiepileptic drugs (e.g., carbamazepine), anti-HIV agents (e.g., ritonavir), antipsychotics (e.g., clozapine), cardiovascular drugs (e.g., procainamide and hydralazine), immunosupressants (e.g., cyclosporine A), inhalational anesthetics (e.g., halothane), nonsteroidal anti-inflammatory drugs (NSAIDSs) (e.g., diclofenac), and steroids and their receptor modulators (e.g., estrogens and tamoxifen). Some herbal and dietary constituents are also bioactivated to reactive metabolites capable of binding covalently and inactivating cytochrome P450s (CYPs). A number of important target proteins of drugs have been identified by mass spectrometric techniques and proteomic approaches. The covalent binding and formation of drug-protein adducts are generally considered to be related to drug toxicity, and selective protein covalent binding by drug metabolites may lead to selective organ toxicity. However, the mechanisms involved in the protein adduct-induced toxicity are largely undefined, although it has been suggested that drug-protein adducts may cause toxicity either through impairing physiological functions of the modified proteins or through immune-mediated mechanisms. In addition, mechanism-based inhibition of CYPs may result in toxic drug-drug interactions. The clinical consequences of drug bioactivation and covalent binding to proteins are unpredictable, depending on many factors that are associated with the administered drugs and patients. Further studies using proteomic and genomic approaches with high throughput capacity are needed to identify the protein targets of reactive drug metabolites, and to elucidate the structure-activity relationships of drug's covalent binding to proteins and their clinical outcomes.
Collapse
Affiliation(s)
- Shufeng Zhou
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
28
|
Karlsson KM, Spoof LEM, Meriluoto JAO. Quantitative LC-ESI-MS analyses of microcystins and nodularin-R in animal tissue--matrix effects and method validation. ENVIRONMENTAL TOXICOLOGY 2005; 20:381-9. [PMID: 15892039 DOI: 10.1002/tox.20115] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The matrix effects and signal response in LC-MS analysis of six microcystins and nodularin-R were studied in mussels and liver samples from the common eider and rainbow trout. The instrumentation used in the study was a triple quadrupole MS with electrospray ionization. The results from the spiked tissue samples showed that both signal suppression and enhancement occurred. The recorded matrix effects were not severe; all studied toxins could be detected with sufficient limit of detection in all matrices. The results indicate, however, that matrix effects must be monitored for accurate quantification of microcystin and nodularin in tissue samples. Matrix effects can be studied with standard additions in the studied matrix, as was done in this study. Solid-phase extraction (SPE) resulted in a lower limit of detection compared to no cleanup in the sample preparation. SPE also prolonged the chromatographic stability. SPE cleanup is therefore strongly recommeded. Also described in this article are the chromatographic and mass spectrometric details of glutathione and cysteine conjugates, which are the detoxification products of the toxins. LC-MS analysis is suitable for detoxification studies of microcystins and nodularins. Cysteine conjugate was identified as the main detoxification product in a mussel sample that was exposed to toxic cyanobacteria in an aquarium.
Collapse
Affiliation(s)
- Krister M Karlsson
- Department of Biochemistry and Pharmacy, Abo Akademi University, Artillerigatan 6, FIN-20521 Turku, Finland.
| | | | | |
Collapse
|
29
|
Stern ST, Bruno MK, Horton RA, Hill DW, Roberts JC, Cohen SD. Contribution of acetaminophen-cysteine to acetaminophen nephrotoxicity II. Possible involvement of the γ-glutamyl cycle. Toxicol Appl Pharmacol 2005; 202:160-71. [PMID: 15629191 DOI: 10.1016/j.taap.2004.06.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Accepted: 06/07/2004] [Indexed: 11/22/2022]
Abstract
Acetaminophen (APAP) nephrotoxicity has been observed both in humans and research animals. Our recent investigations have focused on the possible involvement of glutathione-derived APAP metabolites in APAP nephrotoxicity and have demonstrated that administration of acetaminophen-cysteine (APAP-CYS) potentiated APAP-induced renal injury with no effects on APAP-induced liver injury. Additionally, APAP-CYS treatment alone resulted in a dose-responsive renal GSH depletion. This APAP-CYS-induced renal GSH depletion could interfere with intrarenal detoxification of APAP or its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI) and may be the mechanism responsible for the potentiation of APAP nephrotoxicity. Renal-specific GSH depletion has been demonstrated in mice and rats following administration of amino acid gamma-glutamyl acceptor substrates for gamma-glutamyl transpeptidase (gamma-GT). The present study sought to determine if APAP-CYS-induced renal glutathione depletion is the result of disruption of the gamma-glutamyl cycle through interaction with gamma-GT. The results confirmed that APAP-CYS-induced renal GSH depletion was antagonized by the gamma-glutamyl transpeptidase (gamma-GT) inhibitor acivicin. In vitro analysis demonstrated that APAP-CYS is a gamma-glutamyl acceptor for both murine and bovine renal gamma-GT. Analysis of urine from mice pretreated with acivicin and then treated with APAP, APAP-CYS, or acetaminophen-glutathione identified a gamma-glutamyl-cysteinyl-acetaminophen metabolite. These findings are consistent with the hypothesis that APAP-CYS contributes to APAP nephrotoxicity by depletion of renal GSH stores through interaction with the gamma-glutamyl cycle.
Collapse
Affiliation(s)
- Stephan T Stern
- Toxicology Program, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06268, USA
| | | | | | | | | | | |
Collapse
|
30
|
Nolin TD, Frye RF, Matzke GR. Hepatic drug metabolism and transport in patients with kidney disease. Am J Kidney Dis 2003; 42:906-25. [PMID: 14582035 DOI: 10.1016/j.ajkd.2003.07.019] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The disposition of many drugs is altered in patients with acute (AKD) and chronic kidney disease (CKD). A decline in renal clearance of several drugs has been correlated significantly with residual renal function (ie, creatinine clearance) of subjects. Reductions in nonrenal clearance of some compounds also have been reported and associated with clearance of markers of oxidative and/or conjugative metabolism or P-glycoprotein-mediated transport. Although initial accounts of reduced hepatic microsomal cytochrome P-450 (CYP) content and activity in animal models of AKD and CKD were published almost 25 years ago, it is only in the last decade that technical advances in molecular biology and clinical pharmacology have enabled researchers to begin to characterize the phenotypic expression of individual enzymes and, importantly, distinguish the molecular and/or genetic basis for these changes. The selective modulation of hepatic CYP enzyme activity observed in kidney disease is caused, at least in part, by differentially altered expression of several CYP isoforms. This review summarizes data available through June 2003 regarding the effect of AKD and CKD on drug metabolism. Knowledge of the impact and nature of these alterations associated with kidney disease may facilitate the individualization of medication management in this patient population.
Collapse
Affiliation(s)
- Thomas D Nolin
- Department of Pharmacy Services and Division of Nephrology and Renal Transplantation, Maine Medical Center, Portland, ME, USA
| | | | | |
Collapse
|
31
|
Ross MK, Pegram RA. [35S]-labeling of the Salmonella typhimurium glutathione pool to assess glutathione-mediated DNA binding by 1,2-dibromoethane. Chem Biol Interact 2003; 146:39-49. [PMID: 12902151 DOI: 10.1016/s0009-2797(03)00071-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biotransformation of drugs and environmental chemicals to reactive intermediates is often studied with the use of radiolabeled compounds that are synthesized by expensive and technically difficult procedures. In general, glutathione (GSH) conjugation serves as a detoxification mechanism, and conjugation of reactive intermediates with GSH is often a surrogate marker of reactive species formation. However, several halogenated alkanes can be bioactivated by GSH to yield highly reactive GSH conjugates, some of which are DNA-reactive (e.g. conjugates of 1,2-dibromoethane). The purpose of this study was to metabolically radiolabel the in vivo GSH pool of Salmonella typhimurium with a [35S]-label and to examine the GSH-mediated bioactivation of a model haloalkane, 1,2-dibromoethane, by measuring the binding of [35S]-label to DNA. The strain of Salmonella used in this study had been transformed previously with the gene that codes for rat glutathione transferase theta 1-1 (GSTT1-1), an enzyme that can catalyze formation of genotoxic GSH conjugates. Bacteria were grown to mid-log phase and then incubated with [35S]-L-cysteine in minimal medium (thio-free) until stationary phase of growth was reached. At this stage, the specific activity of Salmonella GSH was estimated to be 7.1 mCi/mmol by derivatization and subsequent HPLC analysis, and GSTT1-1 enzyme activity was still demonstrable in Salmonella cytosol following growth in a minimal medium. The [35S]-labeled bacteria were then exposed to 1,2-dibromoethane (1 mM), and the Salmonella DNA was subsequently purified to quantify [35S]-binding to DNA. The amount of [35S]-label that was covalently bound to DNA in the GSTT1-1-expressing Salmonella strain (33.2 nmol/mg DNA) was sevenfold greater than that of the control strain that does not express GSTT1-1. Neutral thermal hydrolysis of the DNA yielded a single [35S]-labeled adduct with a similar t(R) as S-[2-(N(7)-guanyl)ethyl]GSH, following HPLC analysis of the hydrolysate. This adduct accounted for 95% of the total [35S]-label bound to DNA. Thus, this [35S]-radiolabeling protocol may prove useful for studying the DNA reactivity of GSH conjugates of other halogenated alkanes in a cellular context that maintains GSH at normal physiological levels. This is also, to our knowledge, the first demonstration of de novo incorporation of [35S]-L-cysteine into the bacterial GSH pool.
Collapse
Affiliation(s)
- Matthew K Ross
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
32
|
Rinaldi R, Eliasson E, Swedmark S, Morgenstern R. Reactive intermediates and the dynamics of glutathione transferases. Drug Metab Dispos 2002; 30:1053-8. [PMID: 12228179 DOI: 10.1124/dmd.30.10.1053] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Reactive intermediates are a continuous burden in biology and several defense mechanisms have evolved. Here we focus on the functions of glutathione transferases (GSTs) with the aim to discuss the quantitative aspects of defense against reactive intermediates. Humans excrete approximately 0.1 mmol of thioether conjugates per day. As the amount of GST active sites in liver is approximately 0.5 mmol, it appears that glutathione transferase catalysts are present in tremendous excess. In fact, the known catalytic properties of GSTs reveal that the enzymes can empty the liver glutathione (GSH) pool in a matter of seconds when provided with a suitable substrate. However, based on the urinary output of conjugates (or derivatives thereof), individual GSTs turn over (i.e., catalyze a single reaction) only once every few days. Glutathione transferase overcapacity reflects the fact that there is a linear relation between GST enzyme amount and protection level (provided that GSH is not depleted). Put in a different perspective, a few reactive molecules will always escape conjugation and reach cellular targets. It is therefore not surprising that signaling systems sensing reactive intermediates have evolved resulting in the increase of GSH and GST levels. Precisely for this reason, more moderately reactive electrophiles (Michael acceptors) are receiving growing interest due to their anticarcinogenic properties. Another putative regulatory mechanism involves direct activation of microsomal GST1 by thiol-reactive electrophiles through cysteine 49. The toxicological significance of low levels of reactive intermediates are of interest also in drug development, and here we discuss the use of microsomal GST1 activation as a surrogate detection marker.
Collapse
Affiliation(s)
- Rosanna Rinaldi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
33
|
Kretz O, Guenat C, Beilstein P, Gross G. Coincubation of tissue slices, a new way to study metabolic cooperation between organs: Hepatorenal cooperation in the biotransformation of CGP 47 969 A. J Pharmacol Toxicol Methods 2002; 48:119-26. [PMID: 14565569 DOI: 10.1016/s1056-8719(03)00039-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION There is limited information on in vitro/ex vivo tools to be used for studying interorgan metabolic cooperation. We report here the use of the tissue slice technique for this purpose. METHODS Rat liver and kidney slices were used to study metabolic cooperation for the metabolism of CGP 47 969, a potential anti-inflammatory compound which in vivo is extensively conjugated with glutathione and subsequently degraded via the mercapturic acid pathway. RESULTS Upon incubation with liver slices, CGP 47 969 was extensively conjugated with GSH whilst degradation of the GSH conjugate was moderate. Upon incubation with kidney slices, conjugation of CGP 47 969 with GSH was moderate but degradation of the GSH conjugate was complete. Upon coincubation of CGP 47 969 with liver and kidney slices, both conjugation with GSH and its subsequent degradation were almost complete. Thus, coincubation of liver and kidney slices permitted the efficient in vitro reproduction of the complete biotransformation of CGP 47 969 via its GSH conjugate to the ultimate mercapturic acid metabolite in a one step procedure. DISCUSSION This novel slice coincubation culture could serve as an in vitro model for interorgan cooperation in multistep metabolic processing.
Collapse
Affiliation(s)
- O Kretz
- Novartis Pharma, PCS-Europe ADME, WKL-135.4.24, CH-4002 Basel, Switzerland.
| | | | | | | |
Collapse
|