1
|
Li W, Hu Z, Sun C, Wang Y, Li W, Peng Y, Zheng J. A Metabolic Activation-Based Chemoproteomic Platform to Profile Adducted Proteins Derived from Furan-Containing Compounds. ACS Chem Biol 2022; 17:873-882. [PMID: 35353477 DOI: 10.1021/acschembio.1c00917] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human exposure to widespread furan-containing compounds (FCCs) has drawn much attention due to the high risk of their toxicities. Identifying adducted proteins resulting from the metabolic activation of FCCs is the core to learning the mechanism of FCCs' toxic action. We succeeded in establishing a metabolic activation-based chemoproteomic platform to map FCC-derived protein adducts in cultured primary hepatocytes treated with FCCs and to pinpoint the modification sites, using click chemistry but without alkynylation or azidation of FCCs to be investigated. The proposed platform was systematically verified by biomimetic synthesis, liver microsomal incubation, and primary hepatocyte culture. A mixture of furan, 2-methylfuran, and 2,5-dimethylfuran as model was tested by use of the established platform. A total of hepatic 171 lysine-based adducted proteins and 145 cysteine-based adducted proteins by the reactive metabolites of the three FCCs were enriched and identified (Byonic score ≥ 100). The target proteins were found to mainly participate in ATP synthesis. The technique was also successfully applied to furan-containing natural products. The established platform made it possible to profile covalently adducted proteins, because of potential exposure to a vast inventory of over two million of FCCs documented.
Collapse
Affiliation(s)
- Wei Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Zixia Hu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Chen Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Yuwei Wang
- Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Weiwei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
- Key Laboratory of Environmental Pollution, Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, P. R. China
| |
Collapse
|
2
|
Schleiff MA, Flynn NR, Payakachat S, Schleiff BM, Pinson AO, Province DW, Swamidass SJ, Boysen G, Miller GP. Significance of Multiple Bioactivation Pathways for Meclofenamate as Revealed through Modeling and Reaction Kinetics. Drug Metab Dispos 2020; 49:133-141. [PMID: 33239334 PMCID: PMC7841419 DOI: 10.1124/dmd.120.000254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022] Open
Abstract
Meclofenamate is a nonsteroidal anti-inflammatory drug used in the treatment of mild-to-moderate pain yet poses a rare risk of hepatotoxicity through an unknown mechanism. Nonsteroidal anti-inflammatory drug (NSAID) bioactivation is a common molecular initiating event for hepatotoxicity. Thus, we hypothesized a similar mechanism for meclofenamate and leveraged computational and experimental approaches to identify and characterize its bioactivation. Analyses employing our XenoNet model indicated possible pathways to meclofenamate bioactivation into 19 reactive metabolites subsequently trapped into glutathione adducts. We describe the first reported bioactivation kinetics for meclofenamate and relative importance of those pathways using human liver microsomes. The findings validated only four of the many bioactivation pathways predicted by modeling. For experimental studies, dansyl glutathione was a critical trap for reactive quinone metabolites and provided a way to characterize adduct structures by mass spectrometry and quantitate yields during reactions. Of the four quinone adducts, we were able to characterize structures for three of them. Based on kinetics, the most efficient bioactivation pathway led to the monohydroxy para-quinone-imine followed by the dechloro-ortho-quinone-imine. Two very inefficient pathways led to the dihydroxy ortho-quinone and a likely multiply adducted quinone. When taken together, bioactivation pathways for meclofenamate accounted for approximately 13% of total metabolism. In sum, XenoNet facilitated prediction of reactive metabolite structures, whereas quantitative experimental studies provided a tractable approach to validate actual bioactivation pathways for meclofenamate. Our results provide a foundation for assessing reactive metabolite load more accurately for future comparative studies with other NSAIDs and drugs in general.
Collapse
Affiliation(s)
- Mary Alexandra Schleiff
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Noah R Flynn
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Sasin Payakachat
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Benjamin Mark Schleiff
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Anna O Pinson
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Dennis W Province
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - S Joshua Swamidass
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Gunnar Boysen
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Grover P Miller
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| |
Collapse
|
3
|
Jiang D, Shen M, Ahiadu B, Rusling JF. Organ-Specific Screening for Protein Damage Using Magnetic Bead Bioreactors and LC-MS/MS. Anal Chem 2020; 92:5337-5345. [PMID: 32176468 PMCID: PMC7509849 DOI: 10.1021/acs.analchem.9b05871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A new 96-well plate methodology for fast, enzyme-multiplexed screening for metabolite-protein adducts was developed. Magnetic beads coated with metabolic enzymes were used to make potentially reactive metabolites that can react with test protein in the wells, followed by sample workup in multiple 96-well filter plates for LC-MS/MS analysis. Incorporation of human microsomes from multiple organs and selected supersomes of single cytochrome P450 (cyt P450) enzymes on the magnetic beads provided a broad spectrum of metabolic enzymes. The reacted protein was then isolated, denatured, reduced, alkylated, and digested, and peptides were collected in a sequence of 96-well filter plates for analysis. Method performance was evaluated by trapping acetaminophen reactive metabolite N-acetyl-p-benzoquinoneimine (NAPQI) with human glutathione S-transferase pi (hGSTP), human serum albumin (HSA), and bovine serum albumin (BSA) as model target proteins. Relative amounts of acetaminophen metabolite and hGSTP adducts were compared with 10 different cyt P450 enzymes. Human liver microsomes and CYP1A2 supersomes showed the highest bioactivation rate for adduct formation, in which all four cysteines of hGSTP reacted with NAPQI. Eight cysteines of HSA and four cysteines of BSA have been detected to react with NAPQI. This method has the potential for fast multienzyme protein adduct screening with high efficiency and accuracy.
Collapse
Affiliation(s)
- Di Jiang
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Min Shen
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ben Ahiadu
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - James F Rusling
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Surgery and Neag Cancer Center, UConn Health, Farmington, Connecticut 06032, United States
- Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- School of Chemistry, National University of Ireland at Galway, Galway H91 TK33, Ireland
| |
Collapse
|
4
|
Geib T, Lento C, Wilson DJ, Sleno L. Liquid Chromatography-Tandem Mass Spectrometry Analysis of Acetaminophen Covalent Binding to Glutathione S-Transferases. Front Chem 2019; 7:558. [PMID: 31457004 PMCID: PMC6700392 DOI: 10.3389/fchem.2019.00558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/22/2019] [Indexed: 01/12/2023] Open
Abstract
Acetaminophen (APAP)-induced hepatotoxicity is the most common cause of acute liver failure in the Western world. APAP is bioactivated to N-acetyl p-benzoquinone imine (NAPQI), a reactive metabolite, which can subsequently covalently bind to glutathione and protein thiols. In this study, we have used liquid chromatography-tandem mass spectrometry (LC-MS/MS) to characterize NAPQI binding to human glutathione S-transferases (GSTs) in vitro. GSTs play a crucial role in the detoxification of reactive metabolites and therefore are interesting target proteins to study in the context of APAP covalent binding. Recombinantly-expressed and purified GSTs were used to assess NAPQI binding in vitro. APAP biotransformation to NAPQI was achieved using rat liver microsomes or human cytochrome P450 Supersomes in the presence of GSTA1, M1, M2, or P1. Resulting adducts were analyzed using bottom-up proteomics, with or without LC fractionation prior to LC-MS/MS analysis on a quadrupole-time-of-flight instrument with data-dependent acquisition (DDA). Targeted methods using multiple reaction monitoring (MRM) on a triple quadrupole platform were also developed by quantitatively labeling all available cysteine residues with a labeling reagent yielding isomerically-modified peptides following enzymatic digestion. Seven modified cysteine sites were confirmed, including Cys112 in GSTA1, Cys78 in GSTM1, Cys115 and 174 in GSTM2, as well as Cys15, 48, and 170 in GSTP1. Most modified peptides could be detected using both untargeted (DDA) and targeted (MRM) approaches, however the latter yielded better detection sensitivity with higher signal-to-noise and two sites were uniquely found by MRM.
Collapse
Affiliation(s)
- Timon Geib
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
| | - Cristina Lento
- Department of Chemistry, The Centre for Research in Mass Spectrometry, York University, Toronto, ON, Canada
| | - Derek J Wilson
- Department of Chemistry, The Centre for Research in Mass Spectrometry, York University, Toronto, ON, Canada
| | - Lekha Sleno
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
| |
Collapse
|
5
|
Kondyurin A, Lau K, Tang F, Akhavan B, Chrzanowski W, Lord MS, Rnjak-Kovacina J, Bilek MM. Plasma Ion Implantation of Silk Biomaterials Enabling Direct Covalent Immobilization of Bioactive Agents for Enhanced Cellular Responses. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17605-17616. [PMID: 29733628 DOI: 10.1021/acsami.8b03182] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Silk fibroin isolated from Bombyx mori cocoons is a promising material for a range of biomedical applications, but it has no inherent cell-interactive domains, necessitating functionalization with bioactive molecules. Here we demonstrate significantly enhanced cell interactions with silk fibroin biomaterials in the absence of biofunctionalization following surface modification using plasma immersion ion implantation (PIII). Further, PIII treated silk fibroin biomaterials supported direct covalent immobilization of proteins on the material surface in the absence of chemical cross-linkers. Surface analysis after nitrogen plasma and PIII treatment at 20 kV revealed that the silk macromolecules are significantly fragmented, and at the higher fluences of implanted ions, surface carbonization was observed to depths corresponding to that of the ion penetration. Consistent with the activity of radicals created in the treated surface layer, oxidation was observed on contact with atmospheric oxygen and the PIII treated surfaces were capable of direct covalent immobilization of bioactive macromolecules. Changes in thickness, amide and nitrile groups, refractive index, and extinction coefficient in the wavelength range 400-1000 nm as a function of ion fluence are presented. Reactions responsible for the restructuring of the silk surface under ion beam treatment that facilitate covalent binding of proteins and a significant improvement in cell interactions on the modified surface are proposed.
Collapse
Affiliation(s)
- Alexey Kondyurin
- School of Physics , University of Sydney , Sydney NSW 2006 , Australia
| | - Kieran Lau
- Graduate School of Biomedical Engineering , University of New South Wales , Sydney NSW 2052 , Australia
| | - Fengying Tang
- Graduate School of Biomedical Engineering , University of New South Wales , Sydney NSW 2052 , Australia
| | - Behnam Akhavan
- School of Physics , University of Sydney , Sydney NSW 2006 , Australia
- School of Aerospace, Mechanical and Mechatronic Engineering , University of Sydney , Sydney NSW 2006 , Australia
| | - Wojciech Chrzanowski
- School of Pharmacy , University of Sydney , Sydney NSW 2006 , Australia
- University of Sydney Nano Institute , University of Sydney , Sydney NSW 2006 , Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering , University of New South Wales , Sydney NSW 2052 , Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering , University of New South Wales , Sydney NSW 2052 , Australia
| | - Marcela M Bilek
- School of Physics , University of Sydney , Sydney NSW 2006 , Australia
- School of Aerospace, Mechanical and Mechatronic Engineering , University of Sydney , Sydney NSW 2006 , Australia
- University of Sydney Nano Institute , University of Sydney , Sydney NSW 2006 , Australia
- Charles Perkins Centre , University of Sydney , Sydney NSW 2006 , Australia
| |
Collapse
|
6
|
Whitby LR, Obach RS, Simon GM, Hayward MM, Cravatt BF. Quantitative Chemical Proteomic Profiling of the in Vivo Targets of Reactive Drug Metabolites. ACS Chem Biol 2017. [PMID: 28636309 DOI: 10.1021/acschembio.7b00346] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Idiosyncratic liver toxicity represents an important problem in drug research and pharmacotherapy. Reactive drug metabolites that modify proteins are thought to be a principal factor in drug-induced liver injury. Here, we describe a quantitative chemical proteomic method to identify the targets of reactive drug metabolites in vivo. Treating mice with clickable analogues of four representative hepatotoxic drugs, we demonstrate extensive covalent binding that is confined primarily to the liver. Each drug exhibited a distinct target profile that, in certain cases, showed strong enrichment for specific metabolic pathways (e.g., lipid/sterol pathways for troglitazone). Site-specific proteomics revealed that acetaminophen reacts with high stoichiometry with several conserved, functional (seleno)cysteine residues throughout the liver proteome. Our findings thus provide an advanced experimental framework to characterize the proteomic reactivity of drug metabolites in vivo, revealing target profiles that may help to explain mechanisms and identify risk factors for drug-induced liver injury.
Collapse
Affiliation(s)
- Landon R. Whitby
- The
Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92307, United States
| | - R. Scott Obach
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gabriel M. Simon
- Vividion Therapeutics, 3033 Science
Park Rd Suite D, San Diego, California 92121, United States
| | - Matthew M. Hayward
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Benjamin F. Cravatt
- The
Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92307, United States
| |
Collapse
|
7
|
Kondyurina I, Wise SG, Ngo AKY, Filipe EC, Kondyurin A, Weiss AS, Bao S, Bilek MMM. Plasma mediated protein immobilisation enhances the vascular compatibility of polyurethane with tissue matched mechanical properties. Biomed Mater 2017; 12:045002. [DOI: 10.1088/1748-605x/aa6eb6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
8
|
Gan J, Zhang H, Humphreys WG. Drug–Protein Adducts: Chemistry, Mechanisms of Toxicity, and Methods of Characterization. Chem Res Toxicol 2016; 29:2040-2057. [DOI: 10.1021/acs.chemrestox.6b00274] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jinping Gan
- Department of Biotransformation, Bristol-Myers Squibb Pharmaceutical Company, Princeton, New Jersey 08540, United States
| | - Haiying Zhang
- Department of Biotransformation, Bristol-Myers Squibb Pharmaceutical Company, Princeton, New Jersey 08540, United States
| | - W. Griffith Humphreys
- Department of Biotransformation, Bristol-Myers Squibb Pharmaceutical Company, Princeton, New Jersey 08540, United States
| |
Collapse
|
9
|
Golizeh M, Geib T, Sleno L. Identification of 4-hydroxynonenal protein targets in rat, mouse and human liver microsomes by two-dimensional liquid chromatography/tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:1488-1494. [PMID: 27321836 DOI: 10.1002/rcm.7577] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 06/06/2023]
Abstract
RATIONALE 4-Hydroxynonenal (HNE), endogenously generated through peroxidation and breakdown of polyunsaturated fatty acids, has been linked to a number of adverse biological effects through carbonylation of essential biomolecules. Covalent binding of HNE to proteins can alter their structure and functions, causing cell damage as well as adverse immune responses. The liver plays a predominant role in metabolic transformations and hepatic proteins are often targeted by reactive metabolites. METHODS Rat, mouse and human liver microsomes were incubated with HNE, enzymatically digested, and subjected to strong cation-exchange peptide fractionation prior to liquid chromatography/tandem mass spectrometry (LC/MS/MS) analysis coupled to electrospray ionization quadrupole time-of-flight (QqTOF) mass spectrometry. HNE-modified peptides were detected by probability-driven peptide spectral matching and comparative analysis between treated and control samples, and confirmed based on accurate mass and high-resolution MS/MS spectra. RESULTS A total of 99, 123 and 51 HNE-modified peptides were identified in rat, mouse and human liver microsomes related to 76, 103 and 44 target proteins, respectively. Eight proteins were found to be adducted by HNE in all three species, including ATP synthase, carbamoyl phosphate synthase, cytochrome P450 1A2, glutamate dehydrogenase 1, protein ERGIC-53, protein disulfide-isomerase, and voltage-dependent anion-selective channel protein 1. These proteins play crucial roles in cellular processes and their covalent modification could potentially alter their function and lead to cytotoxicity. CONCLUSIONS An analytical approach was developed for the identification of in vitro HNE protein targets in rat, mouse and human liver microsomes using two-dimensional (2D) LC/MS/MS. This approach can be applied to study HNE modification of proteins in vitro and in vivo, providing insight into the toxicology of HNE protein adduction. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Makan Golizeh
- Chemistry Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, QC, Canada
| | - Timon Geib
- Chemistry Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, QC, Canada
| | - Lekha Sleno
- Chemistry Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, QC, Canada
| |
Collapse
|
10
|
Golizeh M, LeBlanc A, Sleno L. Identification of Acetaminophen Adducts of Rat Liver Microsomal Proteins using 2D-LC-MS/MS. Chem Res Toxicol 2015; 28:2142-50. [DOI: 10.1021/acs.chemrestox.5b00317] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Makan Golizeh
- Chemistry
Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, Québec H2X 2J6, Canada
| | - André LeBlanc
- Chemistry
Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, Québec H2X 2J6, Canada
| | - Lekha Sleno
- Chemistry
Department/Pharmaqam, Université du Québec à Montréal (UQÀM), Montréal, Québec H2X 2J6, Canada
| |
Collapse
|
11
|
Zhang H, Gan J, Shu YZ, Humphreys WG. High-Resolution Mass Spectrometry-Based Background Subtraction for Identifying Protein Modifications in a Complex Biological System: Detection of Acetaminophen-Bound Microsomal Proteins Including Argininosuccinate Synthetase. Chem Res Toxicol 2015; 28:775-81. [DOI: 10.1021/tx500526s] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Haiying Zhang
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - Jinping Gan
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - Yue-Zhong Shu
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| | - W. Griffith Humphreys
- Biotransformation, Bristol-Myers Squibb Research and Development, Princeton, New Jersey 08543, United States
| |
Collapse
|
12
|
Yang Y, Xiao Q, Humphreys WG, Dongre A, Shu YZ. Identification of Human Liver Microsomal Proteins Adducted by a Reactive Metabolite Using Shotgun Proteomics. Chem Res Toxicol 2014; 27:1537-46. [DOI: 10.1021/tx500181p] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Yanou Yang
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Qing Xiao
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - W. Griffith Humphreys
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Ashok Dongre
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| | - Yue-Zhong Shu
- Bristol-Myers Squibb Research and Development, 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, United States
| |
Collapse
|
13
|
Identification and quantification of drug-albumin adducts in serum samples from a drug exposure study in mice. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 917-918:53-61. [PMID: 23353939 DOI: 10.1016/j.jchromb.2012.12.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 01/11/2023]
Abstract
The formation of drug-protein adducts following the bioactivation of drugs to reactive metabolites has been linked to adverse drug reactions (ADRs) and is a major complication in drug discovery and development. Identification and quantification of drug-protein adducts in vivo may lead to a better understanding of drug toxicity, but is challenging due to their low abundance in the complex biological samples. Human serum albumin (HSA) is a well-known target of reactive drug metabolites due to the free cysteine on position 34 and is often the first target to be investigated in covalent drug binding studies. Presented here is an optimized strategy for targeted analysis of low-level drug-albumin adducts in serum. This strategy is based on selective extraction of albumin from serum through affinity chromatography, efficient sample treatment and clean-up using gel filtration chromatography followed by tryptic digestion and LC-MS analysis. Quantification of the level of albumin modification was performed through a comparison of non-modified and drug-modified protein based on the relative peak area of the tryptic peptide containing the free cysteine residue. The analysis strategy was applied to serum samples resulting from a drug exposure experiment in mice, which was designed to study the effects of different acetaminophen (APAP) treatments on drug toxicity. APAP is bioactivated to N-acetyl-p-benzoquinoneimine (NAPQI) in both humans and mice and is known to bind to cysteine 34 (cys34) of HSA. Analysis of the mouse serum samples revealed the presence of extremely low-level NAPQI-albumin adducts of approximately 0.2% of the total mouse serum albumin (MSA), regardless of the length of drug exposure. Due to the targeted nature of the strategy, the NAPQI-adduct formation on cys34 could be confirmed while adducts to the second free cysteine on position 579 of MSA were not detected.
Collapse
|
14
|
Hu L, Paul Fawcett J, Gu J. Protein target discovery of drug and its reactive intermediate metabolite by using proteomic strategy. Acta Pharm Sin B 2012. [DOI: 10.1016/j.apsb.2012.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
15
|
Deng Z, Zhong D, Chen X. Identification of modification sites on human serum albumin and human hemoglobin adducts with houttuynin using liquid chromatography coupled with mass spectrometry. Biomed Chromatogr 2012; 26:1377-85. [DOI: 10.1002/bmc.2708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 12/31/2011] [Accepted: 12/31/2011] [Indexed: 11/12/2022]
Affiliation(s)
- Zhipeng Deng
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai; 201203; China
| | - Dafang Zhong
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai; 201203; China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai; 201203; China
| |
Collapse
|
16
|
Moro S, Chipman JK, Antczak P, Turan N, Dekant W, Falciani F, Mally A. Identification and Pathway Mapping of Furan Target Proteins Reveal Mitochondrial Energy Production and Redox Regulation as Critical Targets of Furan Toxicity. Toxicol Sci 2012; 126:336-52. [DOI: 10.1093/toxsci/kfs005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
17
|
Simulation of the oxidative metabolism of diclofenac by electrochemistry/(liquid chromatography/)mass spectrometry. Anal Bioanal Chem 2012; 403:345-54. [DOI: 10.1007/s00216-011-5665-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/07/2011] [Accepted: 12/15/2011] [Indexed: 10/14/2022]
|
18
|
Roth RA, Ganey PE. Animal models of idiosyncratic drug-induced liver injury—Current status. Crit Rev Toxicol 2011; 41:723-39. [DOI: 10.3109/10408444.2011.575765] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
19
|
Switzar L, Giera M, Lingeman H, Irth H, Niessen W. Protein digestion optimization for characterization of drug–protein adducts using response surface modeling. J Chromatogr A 2011; 1218:1715-23. [DOI: 10.1016/j.chroma.2010.12.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/08/2010] [Accepted: 12/11/2010] [Indexed: 10/18/2022]
|
20
|
Hirsh SL, Bilek MMM, Nosworthy NJ, Kondyurin A, dos Remedios CG, McKenzie DR. A comparison of covalent immobilization and physical adsorption of a cellulase enzyme mixture. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:14380-8. [PMID: 20704318 DOI: 10.1021/la1019845] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
This paper reports the first use of a linker-free covalent approach for immobilizing an enzyme mixture. Adsorption from a mixture is difficult to control due to varying kinetics of adsorption, variations in the degree of unfolding and competitive binding effects. We show that surface activation by plasma immersion ion implantation (PIII) produces a mildly hydrophilic surface that covalently couples to protein molecules and avoids these issues, allowing the attachment of a uniform monolayer from a cellulase enzyme mixture. Atomic force microscopy (AFM) showed that the surface layer of the physically adsorbed cellulase layer on the mildly hydrophobic surface (without PIII) consisted of aggregated enzymes that changed conformation with incubation time. The evolution observed is consistent with the existence of transient complexes previously postulated to explain the long time constants for competitive displacement effects in adsorption from enzyme mixtures. AFM indicated that the covalently coupled bound layer to the PIII-treated surface consisted of a stable monolayer without enzyme aggregates, and became a double layer at longer incubation times. Light scattering analysis showed no indication of aggregates in the solution at room temperature, which indicates that the surface without PIII-treatment induced enzyme aggregation. A model for the attachment process of a protein mixture that includes the adsorption kinetics for both surfaces is presented.
Collapse
Affiliation(s)
- S L Hirsh
- Applied and Plasma Physics, School of Physics (A28), The University of Sydney, Sydney, NSW 2006, Australia.
| | | | | | | | | | | |
Collapse
|
21
|
Bax DV, McKenzie DR, Weiss AS, Bilek MM. The linker-free covalent attachment of collagen to plasma immersion ion implantation treated polytetrafluoroethylene and subsequent cell-binding activity. Biomaterials 2010; 31:2526-34. [DOI: 10.1016/j.biomaterials.2009.12.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 12/03/2009] [Indexed: 01/07/2023]
|
22
|
Bax DV, McKenzie DR, Weiss AS, Bilek MM. Linker-free covalent attachment of the extracellular matrix protein tropoelastin to a polymer surface for directed cell spreading. Acta Biomater 2009; 5:3371-81. [PMID: 19463976 DOI: 10.1016/j.actbio.2009.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 04/02/2009] [Accepted: 05/12/2009] [Indexed: 12/28/2022]
Abstract
Polymers are used for the fabrication of many prosthetic implants. It is desirable for these polymers to promote biological function by promoting the adhesion, differentiation and viability of cells. Here we have used plasma immersion ion implantation (PIII) treatment of polystyrene to modify the polymer surface, and so modulate the binding of the extracellular matrix protein tropoelastin. PIII treated, but not untreated polystyrene, bound tropoelastin in a sodium dodecyl sulfate (SDS)-resistant manner, consistent with previous enzyme-binding data that demonstrated the capability of these surfaces to covalently attach proteins without employing chemical linking molecules. Furthermore sulfo-NHS acetate (SNA) blocking of tropoelastin lysine side chains eliminated the SDS-resistant binding of tropoelastin to PIII-treated polystyrene. This implies tropoelastin is covalently attached to the PIII-treated surface via its lysine side chains. Cell spreading was only observed on tropoelastin coated, PIII-treated polystyrene surfaces, indicating that tropoelastin was more biologically active on the PIII-treated surface compared to the untreated surface. A contact mask was used to pattern the PIII treatment. Following tropoelastin attachment, cells spread preferentially on the PIII-treated sections of the polystyrene surface. This demonstrates that PIII treatment of polystyrene improves the polymer's tropoelastin binding properties, with advantages for tissue engineering and prosthetic design.
Collapse
|
23
|
Witzmann FA, Richardson MR. Two-dimensional gels for toxicological drug discovery applications. Expert Opin Drug Metab Toxicol 2009; 2:103-11. [PMID: 16863472 DOI: 10.1517/17425255.2.1.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Two-dimensional gel electrophoresis (2DGE) continues to be a useful approach to study protein expression. Although liquid chromatographic and mass spectrometric approaches that overcome some of the limitations and labour intensity of 2DGE are increasingly popular, this electrophoretic approach still has exceptional relevance in toxicology. Despite the technical challenges, pharmacologists/toxicologists continue to use gel-based proteomics to assess the biological and health effects of chemical treatment and exposure. This brief review addresses the use of 2DGE-based proteomics in drug development and toxicology, emphasising its unique strengths and weaknesses, and considers recent developments in this strategy that have evolved to directly confront the issues of dynamic range and reproducibility that have previously limited the overall use of 2D electrophoresis.
Collapse
Affiliation(s)
- Frank A Witzmann
- Indiana University School of Medicine, Department of Cellular & Integrative Physiology, Biotechnology & Research Training Center, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
24
|
Tzouros M, Pähler A. A Targeted Proteomics Approach to the Identification of Peptides Modified by Reactive Metabolites. Chem Res Toxicol 2009; 22:853-62. [DOI: 10.1021/tx800426x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Manuel Tzouros
- Drug Metabolism and Pharmacokinetics, Non-Clinical Safety, Pharmaceuticals Division, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Axel Pähler
- Drug Metabolism and Pharmacokinetics, Non-Clinical Safety, Pharmaceuticals Division, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| |
Collapse
|
25
|
Ikegawa S, Yamamoto T, Miyashita T, Okihara R, Ishiwata S, Sakai T, Chong RH, Maeda M, Hofmann AF, Mitamura K. Production and characterization of a monoclonal antibody to capture proteins tagged with lithocholic acid. ANAL SCI 2008; 24:1475-80. [PMID: 18997378 DOI: 10.2116/analsci.24.1475] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Reactive metabolic-modified proteins have been proposed to play an important role in the mechanism(s) of the hepatotoxicity and colon cancer of lithocholic acid (LCA). To identify cellular proteins chemically modified with LCA, we have generated a monoclonal antibody that recognizes the 3alpha-hydroxy-5beta-steroid moiety of LCA. The spleen cells from a BALB/c mouse, which was immunized with an immunogen in which the side chain of LCA was coupled to bovine serum albumin (BSA) via a succinic acid spacer, was fused with SP2/0 myeloma cells to generate antibody-secreting hybridoma clones. The resulting monoclonal antibody (gamma2b, kappa) was specific to LCA-N(alpha)-BOC-lysine as well as the amidated and nonamidated forms of LCA. The immunoblot enabled the detection of LCA residues anchored on BSA and lysozyme. The antibody will be useful for monitoring the generation, localization, and capture of proteins tagged with LCA, which may be the cause of LCA-induced toxicity.
Collapse
Affiliation(s)
- Shigeo Ikegawa
- Faculty of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-osaka 577-8502, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lohmann W, Hayen H, Karst U. Covalent Protein Modification by Reactive Drug Metabolites Using Online Electrochemistry/Liquid Chromatography/Mass Spectrometry. Anal Chem 2008; 80:9714-9. [DOI: 10.1021/ac801699g] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Wiebke Lohmann
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| | - Heiko Hayen
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| | - Uwe Karst
- Institut für Anorganische und Analytische Chemie, Westfälische Wilhelms—Universität Münster, Corrensstrasse 30, 48149 Münster, Germany, and ISAS—Institute for Analytical Sciences, Bunsen-Kirchhoff-Strasse 11, 44139 Dortmund, Germany
| |
Collapse
|
27
|
Hoos JS, Damsten MC, de Vlieger JSB, Commandeur JNM, Vermeulen NPE, Niessen WMA, Lingeman H, Irth H. Automated detection of covalent adducts to human serum albumin by immunoaffinity chromatography, on-line solution phase digestion and liquid chromatography–mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 859:147-56. [PMID: 17913598 DOI: 10.1016/j.jchromb.2007.09.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 09/05/2007] [Accepted: 09/12/2007] [Indexed: 01/09/2023]
Abstract
A generic method for the detection of covalent adducts to the cysteine-34 residue of human serum albumin (HSA) has been developed, based on an on-line combination of immunoaffinity chromatography for selective sample pre-treatment, solution phase digestion, liquid chromatography and tandem mass spectrometry. Selective anti-HSA antibodies immobilized on agarose were used for sample pre-concentration and purification of albumin from the chemically produced alkylated HSA. After elution, HSA and HSA adducts are mixed with pronase and directed to a reaction capillary kept at a digestion temperature of 70 degrees C. The digestion products were trapped on-line on a C18 SPE cartridge. The peptides were separated on a reversed-phase column using a gradient of organic modifier and subsequently detected using tandem mass spectrometry. Modified albumin samples consisted of synthetically alkylated HSA by the reactive metabolite of acetaminophen, N-acetyl-p-benzoquinoneimine (NAPQI), and using the alkylating agent 1-chloro-2,4-dinitrobenzene (CDNB) as reference. The resulting mixture of alkylated versus non-modified albumin has been applied to the on-line system, and alkylation of HSA is revealed by the detection of the modified marker tetra-peptide glutamine-cysteine-proline-phenylalanine (QCPF) adducts NAPQI-QCPF and CDNB-QCPF. Detection of alkylated species was enabled by the use of data comparison algorithms to distinguish between unmodified and modified HSA samples. The in-solution digestion proved to be a useful tool for enabling fast (less than 2 min) and reproducible on-line digestion of HSA. A detection limit of 1.5 micromol/L of modified HSA could be obtained by applying 10 microL of NAPQI-HSA sample.
Collapse
Affiliation(s)
- Johannes S Hoos
- Vrije Universiteit Amsterdam, Faculty of Sciences, Section Analytical Chemistry & Applied Spectroscopy, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Lu X, Huang WH, Ai F, Wang ZL, Cheng JK. Indirect determination of pyruvic acid by capillary electrophoresis with amperometric detection. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 857:347-51. [PMID: 17723321 DOI: 10.1016/j.jchromb.2007.07.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 07/24/2007] [Accepted: 07/25/2007] [Indexed: 10/23/2022]
Abstract
A method of indirectly measuring pyruvic acid (PA) by capillary electrophoresis with amperometric detection is proposed for the first time. It is based on the oximation reaction between PA and hydroxylamine (NH(2)OH), and the quantification of PA was performed by direct and sensitive amperometric detection of excessive NH(2)OH after the oximation reaction. This method displayed a good sensitivity, and the detection limits of NH(2)OH and PA are 1.76 x 10(-7) and 3.88 x 10(-7)mol/L, respectively at S/N=3. The linear relationship between the peak current and PA concentration is exhibited over the range from 4 x 10(-6) to 1 x 10(-4)mol/L. This method has been applied to determine PA in rat plasma with satisfactory results.
Collapse
Affiliation(s)
- Xin Lu
- Department of Chemistry, Wuhan University, Wuhan, China
| | | | | | | | | |
Collapse
|
29
|
Damsten MC, Commandeur JNM, Fidder A, Hulst AG, Touw D, Noort D, Vermeulen NPE. Liquid Chromatography/Tandem Mass Spectrometry Detection of Covalent Binding of Acetaminophen to Human Serum Albumin. Drug Metab Dispos 2007; 35:1408-17. [PMID: 17510247 DOI: 10.1124/dmd.106.014233] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Covalent binding of reactive electrophilic intermediates to proteins is considered to play an important role in the processes leading to adverse drug reactions and idiosyncratic drug reactions. Consequently, both for the discovery and the development of new drugs, there is a great interest in sensitive methodologies that enable the detection of covalent binding of drugs and drug candidates in vivo. In this work, we present a strategy for the generation and analysis of drug adducts to human serum albumin. Our methodology is based on the isolation of albumin from blood, its digestion to peptides by pronase E, and the sensitive detection of adducts to the characteristic cysteine-proline-phenylalanine (CPF) tripeptide by liquid chromatography/tandem mass spectrometry. We chose acetaminophen (APAP) as a model compound because this drug is known to induce covalent binding to proteins when bioactivated by cytochromes P450 to its reactive N-acetyl-p-benzoquinoneimine metabolite. First, by microsomal incubations of APAP in presence of CPF and/or intact albumin, in vitro reference adducts were generated to determine the mass spectrometric characteristics of the expected CPF adducts and to confirm their formation on pronase E digestion of the alkylated protein. When applying this methodology to albumin isolated from blood of patients exposed to APAP, we were indeed able to detect the corresponding CPF adducts. Therefore, this strategy could be seen as a potential biomonitoring tool to detect in vivo reactive intermediates of drugs and drug candidates, e.g., in the preclinical and clinical development phase.
Collapse
Affiliation(s)
- Micaela C Damsten
- LACDR, Division of Molecular Toxicology, Department of Pharmacochemistry, Vrije Universiteit, de Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
30
|
Desrivot J, Herrenknecht C, Ponchel G, Garbi N, Prina E, Fournet A, Bories C, Figadère B, Hocquemiller R, Loiseau PM. Antileishmanial 2-substituted quinolines: in vitro behaviour towards biological components. Biomed Pharmacother 2007; 61:441-50. [PMID: 17459651 DOI: 10.1016/j.biopha.2007.03.004] [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: 02/28/2007] [Accepted: 03/09/2007] [Indexed: 11/16/2022] Open
Abstract
Quinolines substituted on their carbon 2 have in vivo antileishmanial activity but some of them could not be detected in plasma when assayed for pharmacokinetic studies, suggesting a sequestration of the drugs by components of the blood compartment. The present study, performed on three quinolines (1, 2 and 3), showed strong affinity for two of them (2 and 3) with red blood cells (RBCs), whereas quinoline 1 did not react with them. This process was saturable, temperature dependant and positively correlated with the in vitro antileishmanial activity of the quinolines. In addition, a rapid and spontaneous reaction with thiol groups was demonstrated for unsaturated quinolines 2 and 3. The reactivity with RBCs could be part of the compounds targeting to the parasite. These results illustrate that derivatives of the quinoline series with similar antileishmanial in vivo activity have different behaviour in the blood compartment.
Collapse
Affiliation(s)
- Julie Desrivot
- Laboratoire de Pharmacognosie et Chimiothérapie Antiparasitaire, Centre d'Etudes Pharmaceutiques, BioCIS UMR 8076, Université Paris-Sud 11, 5 rue J-B Clément, 92290 Châtenay-Malabry, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hanzlik RP, Koen YM, Theertham B, Dong Y, Fang J. The reactive metabolite target protein database (TPDB)--a web-accessible resource. BMC Bioinformatics 2007; 8:95. [PMID: 17367530 PMCID: PMC1832215 DOI: 10.1186/1471-2105-8-95] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 03/16/2007] [Indexed: 11/23/2022] Open
Abstract
Background The toxic effects of many simple organic compounds stem from their biotransformation to chemically reactive metabolites which bind covalently to cellular proteins. To understand the mechanisms of cytotoxic responses it may be important to know which proteins become adducted and whether some may be common targets of multiple toxins. The literature of this field is widely scattered but expanding rapidly, suggesting the need for a comprehensive, searchable database of reactive metabolite target proteins. Description The Reactive Metabolite Target Protein Database (TPDB) is a comprehensive, curated, searchable, documented compilation of publicly available information on the protein targets of reactive metabolites of 18 well-studied chemicals and drugs of known toxicity. TPDB software enables i) string searches for author names and proteins names/synonyms, ii) more complex searches by selecting chemical compound, animal species, target tissue and protein names/synonyms from pull-down menus, and iii) commonality searches over multiple chemicals. Tabulated search results provide information, references and links to other databases. Conclusion The TPDB is a unique on-line compilation of information on the covalent modification of cellular proteins by reactive metabolites of chemicals and drugs. Its comprehensiveness and searchability should facilitate the elucidation of mechanisms of reactive metabolite toxicity. The database is freely available at
Collapse
Affiliation(s)
- Robert P Hanzlik
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Yakov M Koen
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bhargav Theertham
- Bioinformatics Core Facility, University of Kansas, Lawrence, KS 66047, USA
| | - Yinghua Dong
- Bioinformatics Core Facility, University of Kansas, Lawrence, KS 66047, USA
| | - Jianwen Fang
- Bioinformatics Core Facility, University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
32
|
Koen YM, Gogichaeva NV, Alterman MA, Hanzlik RP. A proteomic analysis of bromobenzene reactive metabolite targets in rat liver cytosol in vivo. Chem Res Toxicol 2007; 20:511-9. [PMID: 17305373 DOI: 10.1021/tx6003166] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metabolic activation and protein covalent binding are early and apparently obligatory events in the cytotoxicity of many simple organic chemicals including drugs and natural products. Although much has been learned about the chemistry of reactive metabolite formation and reactivity toward protein nucleophiles, progress in identifying specific protein targets for reactive metabolites of various protoxins has been much slower. We previously reported nine microsomal and three cytosolic proteins as targets for reactive metabolites of bromobenzene in rat liver. These results, and contemporary work by others, indicate that protein covalent binding is not totally random in cells. Moreover, as protein targets for other protoxins were identified, little commonality of target proteins became apparent. In the present work, we used two-dimensional gel electrophoresis to separate liver cytosolic proteins from rats treated with 14C-bromobenzene; 110 of the 836 observed spots contained measurable radioactivity that varied over a 600-fold range of adduct density. Of these 110 spots, in-gel digestion coupled with mass spectrometry identified apparently single proteins in 57 spots. A few other spots clearly contained more than one identifiable protein, and in several cases, the same protein was identified in several spots having different apparent molecular masses and/or pI. Altogether, 33 unique new protein targets for bromobenzene metabolites were identified and compared to those known for acetaminophen, naphthalene, butylated hydroxytoluene, benzene, thiobenzamide, and halothane via a target protein database available at http://tpdb.medchem.ku.edu:8080/protein_database/. With increasing numbers of target proteins becoming known, more commonality in targeting by reactive metabolites from diverse chemical agents may be seen. Such commonality may help to separate toxicologically significant covalent binding events from a background of covalent binding that is toxicologically inconsequential.
Collapse
Affiliation(s)
- Yakov M Koen
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045-7582, USA
| | | | | | | |
Collapse
|
33
|
Bruyneel B, Hoos JS, Smoluch MT, Lingeman H, Niessen WMA, Irth H. Trace Analysis of Proteins Using Postseparation Solution-Phase Digestion and Electrospray Mass Spectrometric Detection of Marker Peptides. Anal Chem 2006; 79:1591-8. [PMID: 17297960 DOI: 10.1021/ac0616761] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Analytical methodologies for the absolute quantitation of proteins typically include a digest step often using trypsin as the proteolytic enzyme. In the majority of cases, off-line and on-line digestion methods are implemented prior to an LC-MS analysis system, requiring a high sequence coverage for unambiguous protein identification. For proteins with a strong overlap in amino acid sequence, e.g., therapeutic proteins and their metabolites, it is essential to separate proteins prior to digestion and the subsequent electrospray mass spectrometry analysis of marker peptides. Here, we present an on-line postcolumn solution-phase digestion methodology that is based on the continuous infusion of the proteolytic enzyme pepsin downstream to the nano C18 reversed-phase column. Proteins are identified based on their retention time in combination with the detection of specific marker peptides formed in the postcolumn digest. The optimization of important parameters such as enzyme concentration, reaction time, and organic modifier concentration is described. We demonstrated that the continuous-flow solution-phase digest method can be coupled on-line to the reversed-phase gradient liquid chromatography separation of proteins. Detection limits obtained for five model proteins, detected as specific marker peptides with m/z values of 300-1000, range from 30 to 90 fmol, with a linear response up to 3 pmol.
Collapse
Affiliation(s)
- B Bruyneel
- Vrije Universiteit Amsterdam, Faculty of Sciences, Department of Analytical Chemistry and Applied Spectroscopy, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
34
|
Ma S, Subramanian R. Detecting and characterizing reactive metabolites by liquid chromatography/tandem mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2006; 41:1121-39. [PMID: 16967439 DOI: 10.1002/jms.1098] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Metabolic activation of a drug leading to reactive metabolite(s) that can covalently modify proteins is considered an initial step that may lead to drug-induced organ toxicities. Characterization of reactive metabolites is critical to designing new drug candidates with an improved toxicological profile. High performance liquid chromatography (HPLC) coupled with mass spectrometry (MS) predominates over all analytical tools used for screening and characterization of reactive metabolites. In this review, a brief description of experimental approaches employed for assessing reactive metabolites is followed by a discussion on the reactivity of acyl glucuronides and acyl coenzyme A thioesters. Techniques for high-throughput screening and quantitation of reactive metabolite formation are also described, along with proteomic approaches used to identify protein targets and modification sites by reactive metabolites. Strategies for dealing with reactive metabolites are reviewed. In conclusion, we discuss the challenges and future needs in this field of research.
Collapse
Affiliation(s)
- Shuguang Ma
- Pharmacokinetics and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | | |
Collapse
|
35
|
Abstract
The biotechnology industry has undergone rapid growth in recent years largely due to the development and success of protein-based therapeutics for a wide range of disorders. Similar to traditional pharmaceuticals, characterization of a therapeutic protein for its physicochemical properties, process monitoring and lot release is crucial. Electrophoresis in the slab-gel format has and continues to be a mainstay of the protein laboratory; and more recently, CE has begun to make significant inroads for protein analysis in industrial settings. This review focuses on the electrophoresis of proteins with an emphasis on protein-based therapeutics in the capillary, slab-gel and to a lesser extent, the microchip format. Reported applications of electrophoresis at several stages of the biopharmaceutical industry covering the period of 2000-2005 will be discussed.
Collapse
Affiliation(s)
- Michael J Little
- Boehringer Ingelheim, Canada, Research & Development, Laval, Quebec, Canada.
| | | | | |
Collapse
|
36
|
Abstract
Systems biology depends on a comprehensive assignment and characterization of the interactions of proteins and polypeptides (functional proteomics) and of other classes of biomolecules in a given organism. High‐capacity screening methods are in place for ligand capture and interaction screening, but a detailed dynamic characterization of molecular interactions under physiological conditions in efficiently separated mixtures with minimal sample consumption is presently provided only by electrophoretic interaction analysis in capillaries, affinity CE (ACE). This has been realized in different fields of biology and analytical chemistry, and the resulting advances and uses of ACE during the last 2.5 years are covered in this review. Dealing with anything from small divalent metal ions to large supramolecular assemblies, the applications of ACE span from low‐affinity binding of broad specificity being exploited in optimizing selectivity, e.g., in enantiomer analysis to miniaturized affinity technologies, e.g., for fast processing immunoassay. Also, approaches that provide detailed quantitative characterization of analyte–ligand interaction for drug, immunoassay, and aptamer development are increasingly important, but various approaches to ACE are more and more generally applied in biological research. In addition, the present overview emphasizes that distinct challenges regarding sensitivity, parallel processing, information‐rich detection, interfacing with MS, analyte recovery, and preparative capabilities remain. This will be addressed by future technological improvements that will ensure continuing new applications of ACE in the years to come.
Collapse
Affiliation(s)
- Christian Schou
- Department of Autoimmunology, Statens Serum Institute, Copenhagen, Denmark
| | | |
Collapse
|
37
|
Yue W, Koen YM, Williams TD, Hanzlik RP. Use of isotopic signatures for mass spectral detection of protein adduction by chemically reactive metabolites of bromobenzene: studies with model proteins. Chem Res Toxicol 2006; 18:1748-54. [PMID: 16300384 DOI: 10.1021/tx050199z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The cytotoxicity of many small organic compounds often apparently derives from their metabolic activation and covalent binding to cellular proteins. It is therefore of considerable interest to be able to determine, for a given protoxin, which metabolites modify which proteins at which sites. Our laboratory has identified more than 45 target proteins for bromobenzene metabolites in liver by peptide mass mapping after two-dimensional electrophoresis. Through all of this work, we have never observed a bromine-containing peptide. We therefore generated model adducted proteins by carbodiimide coupling of Nalpha-acetyl-Ntau-(p-bromophenyl)-L-histidine (1) and Nalpha-acetyl-Nepsilon-(p-bromophenyl)-L-lysine (2) to bovine pancreatic ribonuclease A. For the adducts, RNase-(1)n and RNase-(2)n, mass spectrometry indicated that n = 0-2 and 0-6, respectively. RNase-(2)n was submitted to in-gel and in-solution digestion with trypsin, and the digests were analyzed by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) and liquid chromatgraphy electrospray ionization MS (LC/ESI-MS) and tandem MS (MS/MS). Sequence coverages observed ranged from 67% with only three modified lysines observed using in-gel digestion and MALDI-TOF analysis, to 100% coverage with all 10 lysines observed in both modified and unmodified form using in-solution digestion and LC/ESI-MS. In the mass spectra of all modified peptides up to 2000 Da, the bromine isotope pattern was obvious by visual inspection; for peptides up to 3600 Da, the isotopic signature could be recognized by visual comparison to simulated spectra. The presence of Br-containing adducts was confirmed by MS/MS analysis of selected peptides. The selection of peaks for MS/MS analysis was significantly facilitated by visual recognition of the bromine isotope pattern, even at signal-to-noise ratios of 10 (or lower in favorable cases). These results indicate that stable isotope labeling may have considerable potential for detecting and locating protein adducts of reactive metabolites.
Collapse
Affiliation(s)
- Weimin Yue
- Department of Medicinal Chemistry, University of Kansas, Malott Hall, Lawrence, Kansas 66045, USA
| | | | | | | |
Collapse
|
38
|
Miksík I, Sedláková P, Mikulíková K, Eckhardt A, Cserhati T, Horváth T. Matrices for capillary gel electrophoresis—a brief overview of uncommon gels. Biomed Chromatogr 2006; 20:458-65. [PMID: 16779791 DOI: 10.1002/bmc.640] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This article gives an overview of uncommon replaceable matrices (gels) for capillary gel electrophoresis. This electrophoretic technique is useful mainly for the separation and analysis of biopolymers-nucleic acids and their fragments, and proteins/peptides. Commonly used gels are not reviewed. Those mentioned and discussed here are gels containing saccharides, newly developed acrylamide-based gels and thermoadjustable viscosity polymers, namely triblock copolymers and grafted polyacrylamide.
Collapse
Affiliation(s)
- Ivan Miksík
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague.
| | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Bateman KP, Baker J, Wilke M, Lee J, Leriche T, Seto C, Day S, Chauret N, Ouellet M, Nicoll-Griffith DA. Detection of covalent adducts to cytochrome P450 3A4 using liquid chromatography mass spectrometry. Chem Res Toxicol 2005; 17:1356-61. [PMID: 15487896 DOI: 10.1021/tx0498861] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein covalent labeling can be an undesirable property of compounds being studied in drug discovery programs. Identifying such compounds relies on the use of radiolabeled material, which requires an investment in time and resources not typically expended until later in the discovery process. We describe the detection of covalent adducts to cytochrome P450 3A4, the most abundant and important P450 from a human and drug discovery viewpoint, using liquid chromatography mass spectrometry. The technique is illustrated using L-754,394 and 6',7'-dihydroxybergamottin, two known inhibitors of P450 3A4. Mass spectrometry of the intact apoprotein as well as the adducted protein is demonstrated. Such methodology may provide the means for screening compounds for covalent protein binding without the use of a radiolabel. It also provides direct information about mechanism-based inhibitors in terms of extent, stoichiometry, and nature of the adduct(s) (mass shift). This information may provide a means for understanding the mechanism of covalent labeling earlier in a drug discovery environment.
Collapse
Affiliation(s)
- Kevin P Bateman
- Department of Medicinal Chemistry, Merck Frosst Canada and Co., 16711 Trans Canada Highway, Kirkland, Quebec H9H 3L1, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|