1
|
The Crystal Structure of Mouse Ces2c, a Potential Ortholog of Human CES2, Shows Structural Similarities in Substrate Regulation and Product Release to Human CES1. Int J Mol Sci 2022; 23:ijms232113101. [DOI: 10.3390/ijms232113101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
Abstract
Members of the carboxylesterase 2 (Ces2/CES2) family have been studied intensively with respect to their hydrolytic function on (pro)drugs, whereas their physiological role in lipid and energy metabolism has been realized only within the last few years. Humans have one CES2 gene which is highly expressed in liver, intestine, and kidney. Interestingly, eight homologous Ces2 (Ces2a to Ces2h) genes exist in mice and the individual roles of the corresponding proteins are incompletely understood. Mouse Ces2c (mCes2c) is suggested as potential ortholog of human CES2. Therefore, we aimed at its structural and biophysical characterization. Here, we present the first crystal structure of mCes2c to 2.12 Å resolution. The overall structure of mCes2c resembles that of the human CES1 (hCES1). The core domain adopts an α/β hydrolase-fold with S230, E347, and H459 forming a catalytic triad. Access to the active site is restricted by the cap, the flexible lid, and the regulatory domain. The conserved gate (M417) and switch (F418) residues might have a function in product release similar as suggested for hCES1. Biophysical characterization confirms that mCes2c is a monomer in solution. Thus, this study broadens our understanding of the mammalian carboxylesterase family and assists in delineating the similarities and differences of the different family members.
Collapse
|
2
|
Shervington LA, Ingham O. Investigating the Stability of Six Phenolic TMZ Ester Analogues, Incubated in the Presence of Porcine Liver Esterase and Monitored by HPLC. Molecules 2022; 27:2958. [PMID: 35566308 PMCID: PMC9103334 DOI: 10.3390/molecules27092958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022] Open
Abstract
Previous published data from our group showed the encouraging in vitro activities of six phenolic temozolomide (TMZ) ester analogues (ES8-ES12 and ES14) with up to a five-fold increase in potency compared to TMZ against glioblastoma multiform cell lines and TMZ-resistant O6-methylguanine-DNA methyl transferase (MGMT)-positive primary cells. This study investigated the stabilities of the six phenolic TMZ ester analogues in the presence of porcine liver esterase (PLE) as a hydrolytic enzyme, using high-performance liquid chromatography (HPLC), monitored by a diode-array detector (DAD). Determining the rates of hydrolysis of the esters provided a useful insight into the feasibility of progressing them to the next phase of drug development. Fifty percent of TMZ esters consisting of para nitro, chloro, phenyl and tolyl groups (ES9, ES10, ES12 and ES14) were hydrolysed within the first 4.2 min of PLE exposure, while the TMZ esters consisting of para methoxy and nitrile groups (ES8 and ES11) demonstrated increased stability, with 50% hydrolysis achieved in 7.3 and 13.7 min, respectively. In conclusion, the survival of these phenolic TMZ esters on route to the target site of a brain tumor would be a challenge, mainly due to the undesirable rapid rate of hydrolysis. These findings therefore pose a question regarding the effectiveness of these esters in an in vivo setting.
Collapse
Affiliation(s)
- Leroy A. Shervington
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| | - Oliver Ingham
- Kindeva Drug Delivery, Bakewell Road, Loughborough LE11 5RB, UK;
| |
Collapse
|
3
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
4
|
|
5
|
Liu J, Shang X, Huang S, Xu Y, Lu J, Zhang Y, Liu Z, Wang X. Construction and Characterization of CRISPR/Cas9 Knockout Rat Model of Carboxylesterase 2a Gene. Mol Pharmacol 2021; 100:480-490. [PMID: 34503976 DOI: 10.1124/molpharm.121.000357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022] Open
Abstract
Carboxylesterase (CES) 2, an important metabolic enzyme, plays a critical role in drug biotransformation and lipid metabolism. Although CES2 is very important, few animal models have been generated to study its properties and functions. Rat Ces2 is similar to human CES2A-CES3A-CES4A gene cluster, with highly similar gene structure, function, and substrate. In this report, CRISPR-associated protein-9 (CRISPR/Cas9) technology was first used to knock out rat Ces2a, which is a main subtype of Ces2 mostly distributed in the liver and intestine. This model showed the absence of CES2A protein expression in the liver. Further pharmacokinetic studies of diltiazem, a typical substrate of CES2A, confirmed the loss of function of CES2A both in vivo and in vitro. At the same time, the expression of CES2C and CES2J protein in the liver decreased significantly. The body and liver weight of Ces2a knockout rats also increased, but the food intake did not change. Moreover, the deficiency of Ces2a led to obesity, insulin resistance, and liver fat accumulation, which are consistent with the symptoms of nonalcoholic fatty liver disease (NAFLD). Therefore, this rat model is not only a powerful tool to study drug metabolism mediated by CES2 but also a good disease model to study NAFLD. SIGNIFICANCE STATEMENT: Human carboxylesterase (CES) 2 plays a key role in the first-pass hydrolysis metabolism of most oral prodrugs as well as lipid metabolism. In this study, CRISPR/Cas9 technology was used to knock out Ces2a gene in rats for the first time. This model can be used not only in the study of drug metabolism and pharmacokinetics but also as a disease model of nonalcoholic fatty liver disease (NAFLD) and other metabolic disorders.
Collapse
Affiliation(s)
- Jie Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xuyang Shang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Shengbo Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Jian Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Yuanjin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Zongjun Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.Li., X.S., S.H., Y.X., J.Lu., Y.Z., X.W.); and Department of Cardiology, Central Hospital of Shanghai Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China (Z.L.)
| |
Collapse
|
6
|
Singh A, Gao M, Beck MW. Human carboxylesterases and fluorescent probes to image their activity in live cells. RSC Med Chem 2021; 12:1142-1153. [PMID: 34355180 PMCID: PMC8292992 DOI: 10.1039/d1md00073j] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Human carboxylesterases (CESs) are serine hydrolases that are responsible for the phase I metabolism of an assortment of ester, amide, thioester, carbonate, and carbamate containing drugs. CES activity is known to be influenced by a variety of factors including single nucleotide polymorphisms, alternative splicing, and drug-drug interactions. These different factors contribute to interindividual variability of CES activity which has been demonstrated to influence clinical outcomes among people treated with CES-substrate therapeutics. Detailed exploration of the factors that influence CES activity is emerging as an important area of research. The use of fluorescent probes with live cell imaging techniques can selectively visualize the real-time activity of CESs and have the potential to be useful tools to help reveal the impacts of CES activity variations on human health. This review summarizes the properties of the five known human CESs including factors reported to or that could potentially influence their activity before discussing the design aspects and use considerations of CES fluorescent probes in general in addition to highlighting several well-characterized probes.
Collapse
Affiliation(s)
- Anchal Singh
- Department of Chemistry and Biochemistry, Eastern Illinois University Charleston IL 61920 USA +1 217 581 6227
| | - Mingze Gao
- Department of Biological Sciences, Eastern Illinois University Charleston IL 61920 USA
| | - Michael W Beck
- Department of Chemistry and Biochemistry, Eastern Illinois University Charleston IL 61920 USA +1 217 581 6227
| |
Collapse
|
7
|
Bhatt P, Bhatt K, Huang Y, Lin Z, Chen S. Esterase is a powerful tool for the biodegradation of pyrethroid insecticides. CHEMOSPHERE 2020; 244:125507. [PMID: 31835049 DOI: 10.1016/j.chemosphere.2019.125507] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/28/2019] [Indexed: 06/10/2023]
Abstract
Agricultural and household applications of pyrethroid insecticides have significantly increased residual concentrations in living cells and environments. The enhanced concentration is toxic for living beings. Pyrethroid hydrolase enzyme (pyrethroid catalyzing esterase) regulates pyrethroid degradation, and has been well reported in various organisms (bacteria, fungi, insects and animals). Hydrolysis mechanisms of these esterases are different from others and properly function at factors viz., optimum temperature, pH and physicochemical environment. Active site of the enzyme contains common amino acids that play important role in pyrethroid catalysis. Immobilization technology emphasizes the development of better reusable efficiency of pyrethroid hydrolases to carry out large-scale applications for complete degradation of pyrethroids from the environments. In this review we have attempted to provide insights of pyrethroid-degrading esterases in different living systems along with complete mechanisms.
Collapse
Affiliation(s)
- Pankaj Bhatt
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Kalpana Bhatt
- Department of Botany and Microbiology, Gurukula Kangri University, Haridwar 249404, Uttarakhand, India
| | - Yaohua Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Ziqiu Lin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Shaohua Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China.
| |
Collapse
|
8
|
Di L. The Impact of Carboxylesterases in Drug Metabolism and Pharmacokinetics. Curr Drug Metab 2019; 20:91-102. [PMID: 30129408 PMCID: PMC6635651 DOI: 10.2174/1389200219666180821094502] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Carboxylesterases (CES) play a critical role in catalyzing hydrolysis of esters, amides, carbamates and thioesters, as well as bioconverting prodrugs and soft drugs. The unique tissue distribution of CES enzymes provides great opportunities to design prodrugs or soft drugs for tissue targeting. Marked species differences in CES tissue distribution and catalytic activity are particularly challenging in human translation. METHODS Review and summarization of CES fundamentals and applications in drug discovery and development. RESULTS Human CES1 is one of the most highly expressed drug metabolizing enzymes in the liver, while human intestine only expresses CES2. CES enzymes have moderate to high inter-individual variability and exhibit low to no expression in the fetus, but increase substantially during the first few months of life. The CES genes are highly polymorphic and some CES genetic variants show significant influence on metabolism and clinical outcome of certain drugs. Monkeys appear to be more predictive of human pharmacokinetics for CES substrates than other species. Low risk of clinical drug-drug interaction is anticipated for CES, although they should not be overlooked, particularly interaction with alcohols. CES enzymes are moderately inducible through a number of transcription factors and can be repressed by inflammatory cytokines. CONCLUSION Although significant advances have been made in our understanding of CESs, in vitro - in vivo extrapolation of clearance is still in its infancy and further exploration is needed. In vitro and in vivo tools are continuously being developed to characterize CES substrates and inhibitors.
Collapse
Affiliation(s)
- Li Di
- Pfizer Inc., Eastern Point Road, Groton, Connecticut, CT 06354, United States
| |
Collapse
|
9
|
Timperley CM, Abdollahi M, Al-Amri AS, Baulig A, Benachour D, Borrett V, Cariño FA, Geist M, Gonzalez D, Kane W, Kovarik Z, Martínez-Álvarez R, Fusaro Mourão NM, Neffe S, Raza SK, Rubaylo V, Suárez AG, Takeuchi K, Tang C, Trifirò F, van Straten FM, Vanninen PS, Vučinić S, Zaitsev V, Zafar-Uz-Zaman M, Zina MS, Holen S, Forman JE, Alwan WS, Suri V. Advice on assistance and protection by the Scientific Advisory Board of the Organisation for the Prohibition of Chemical Weapons: Part 2. On preventing and treating health effects from acute, prolonged, and repeated nerve agent exposure, and the identification of medical countermeasures able to reduce or eliminate the longer term health effects of nerve agents. Toxicology 2019; 413:13-23. [DOI: 10.1016/j.tox.2018.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/18/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
|
10
|
Ash T, Debnath T, Sarkar S, Gurey P, Das AK. Exploration of assisting behavior of molecular-MO2 (M = Ti, Zr) reagents towards the detoxication of tabun: A DFT study. Chem Phys Lett 2019. [DOI: 10.1016/j.cplett.2019.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Sialic acid as a target for the development of novel antiangiogenic strategies. Future Med Chem 2018; 10:2835-2854. [PMID: 30539670 DOI: 10.4155/fmc-2018-0298] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Sialic acid is associated with glycoproteins and gangliosides of eukaryotic cells. It regulates various molecular interactions, being implicated in inflammation and cancer, where its expression is regulated by sialyltransferases and sialidases. Angiogenesis, the formation of new capillaries, takes place during inflammation and cancer, and represents the outcome of several interactions occurring at the endothelial surface among angiogenic growth factors, inhibitors, receptors, gangliosides and cell-adhesion molecules. Here, we elaborate on the evidences that many structures involved in angiogenesis are sialylated and that their interactions depend on sialic acid with implications in angiogenesis itself, inflammation and cancer. We also discuss the possibility to exploit sialic acid as a target for the development of novel antiangiogenic drugs.
Collapse
|
12
|
Wang YQ, Weng ZM, Dou TY, Hou J, Wang DD, Ding LL, Zou LW, Yu Y, Chen J, Tang H, Ge GB. Nevadensin is a naturally occurring selective inhibitor of human carboxylesterase 1. Int J Biol Macromol 2018; 120:1944-1954. [DOI: 10.1016/j.ijbiomac.2018.09.178] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/26/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
|
13
|
Human carboxylesterases: a comprehensive review. Acta Pharm Sin B 2018; 8:699-712. [PMID: 30245959 PMCID: PMC6146386 DOI: 10.1016/j.apsb.2018.05.005] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Mammalian carboxylesterases (CEs) are key enzymes from the serine hydrolase superfamily. In the human body, two predominant carboxylesterases (CES1 and CES2) have been identified and extensively studied over the past decade. These two enzymes play crucial roles in the metabolism of a wide variety of endogenous esters, ester-containing drugs and environmental toxicants. The key roles of CES in both human health and xenobiotic metabolism arouse great interest in the discovery of potent CES modulators to regulate endobiotic metabolism or to improve the efficacy of ester drugs. This review covers the structural and catalytic features of CES, tissue distributions, biological functions, genetic polymorphisms, substrate specificities and inhibitor properties of CES1 and CES2, as well as the significance and recent progress on the discovery of CES modulators. The information presented here will help pharmacologists explore the relevance of CES to human diseases or to assign the contribution of certain CES in xenobiotic metabolism. It will also facilitate medicinal chemistry efforts to design prodrugs activated by a given CES isoform, or to develop potent and selective modulators of CES for potential biomedical applications.
Collapse
|
14
|
Abdel-Daim A, Ohura K, Imai T. A novel quantification method for serine hydrolases in cellular expression system using fluorophosphonate-biotin probe. Eur J Pharm Sci 2018; 114:267-274. [DOI: 10.1016/j.ejps.2017.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/17/2017] [Accepted: 12/18/2017] [Indexed: 12/26/2022]
|
15
|
Mathew MP, Tan E, Labonte JW, Shah S, Saeui CT, Liu L, Bhattacharya R, Bovonratwet P, Gray JJ, Yarema KJ. Glycoengineering of Esterase Activity through Metabolic Flux-Based Modulation of Sialic Acid. Chembiochem 2017; 18:1204-1215. [PMID: 28218815 PMCID: PMC5757160 DOI: 10.1002/cbic.201600698] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Indexed: 01/09/2023]
Abstract
This report describes the metabolic glycoengineering (MGE) of intracellular esterase activity in human colon cancer (LS174T) and Chinese hamster ovary (CHO) cells. In silico analysis of carboxylesterases CES1 and CES2 suggested that these enzymes are modified with sialylated N-glycans, which are proposed to stabilize the active multimeric forms of these enzymes. This premise was supported by treating cells with butanolylated ManNAc to increase sialylation, which in turn increased esterase activity. By contrast, hexosamine analogues not targeted to sialic acid biosynthesis (e.g., butanoylated GlcNAc or GalNAc) had minimal impact. Measurement of mRNA and protein confirmed that esterase activity was controlled through glycosylation and not through transcription or translation. Azide-modified ManNAc analogues widely used in MGE also enhanced esterase activity and provided a way to enrich targeted glycoengineered proteins (such as CES2), thereby providing unambiguous evidence that the compounds were converted to sialosides and installed into the glycan structures of esterases as intended. Overall, this study provides a pioneering example of the modulation of intracellular enzyme activity through MGE, which expands the value of this technology from its current status as a labeling strategy and modulator of cell surface biological events.
Collapse
Affiliation(s)
- Mohit P. Mathew
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Elaine Tan
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Jason W. Labonte
- Department of Chemical and Biochemical Engineering The Johns Hopkins University, Baltimore, Maryland, USA
| | - Shivam Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Christopher T. Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Lingshu Liu
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Rahul Bhattacharya
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Patawut Bovonratwet
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
| | - Jeffrey J. Gray
- Department of Chemical and Biochemical Engineering The Johns Hopkins University, Baltimore, Maryland, USA
| | - Kevin J. Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center
- Department of Chemical and Biochemical Engineering The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Wang DD, Zou LW, Jin Q, Hou J, Ge GB, Yang L. Recent progress in the discovery of natural inhibitors against human carboxylesterases. Fitoterapia 2017; 117:84-95. [DOI: 10.1016/j.fitote.2017.01.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/13/2017] [Accepted: 01/21/2017] [Indexed: 01/22/2023]
|
17
|
Lei W, Wang DD, Dou TY, Hou J, Feng L, Yin H, Luo Q, Sun J, Ge GB, Yang L. Assessment of the inhibitory effects of pyrethroids against human carboxylesterases. Toxicol Appl Pharmacol 2017; 321:48-56. [PMID: 28242322 DOI: 10.1016/j.taap.2017.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/12/2017] [Accepted: 02/22/2017] [Indexed: 12/12/2022]
Abstract
Pyrethroids are broad-spectrum insecticides that widely used in many countries, while humans may be exposed to these toxins by drinking or eating pesticide-contaminated foods. This study aimed to investigate the inhibitory effects of six commonly used pyrethroids against two major human carboxylesterases (CES) including CES1 and CES2. Three optical probe substrates for CES1 (DME, BMBT and DMCB) and a fluorescent probe substrate for CES2 (DDAB) were used to characterize the inhibitory effects of these pyrethroids. The results demonstrated that most of the tested pyrethroids showed moderate to weak inhibitory effects against both CES1 and CES2, but deltamethrin displayed strong inhibition towards CES1. The IC50 values of deltamethrin against CES1-mediated BMBT, DME, and DMCB hydrolysis were determined as 1.58μM, 2.39μM, and 3.3μM, respectively. Moreover, deltamethrin was cell membrane permeable and capable of inhibition endogenous CES1 in living cells. Further investigation revealed that deltamethrin inhibited CES1-mediated BMBT hydrolysis via competitive manner but noncompetitively inhibited DME or DMCB hydrolysis. The inhibition behaviors of deltamethrin against CES1 were also studied by molecular docking simulation. The results demonstrated that CES1 had at least two different ligand-binding sites, one was the DME site and another was the BMBT site which was identical to the binding site of deltamethrin. In summary, deltamethrin was a strong reversible inhibitor against CES1 and it could tightly bind on CES1 at the same ligand-binding site as BMBT. These findings are helpful for the deep understanding of the interactions between xenobiotics and CES1.
Collapse
Affiliation(s)
- Wei Lei
- The Second Affiliated Hospital of Dalian Medical University, Dalian 110623, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Dan-Dan Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Tong-Yi Dou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jie Hou
- Dalian Medical University, Dalian 116044, China
| | - Liang Feng
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Heng Yin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Qun Luo
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100080, China
| | - Jie Sun
- The Second Affiliated Hospital of Dalian Medical University, Dalian 110623, China
| | - Guang-Bo Ge
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Ling Yang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
18
|
Masson P, Lushchekina SV. Emergence of catalytic bioscavengers against organophosphorus agents. Chem Biol Interact 2016; 259:319-326. [DOI: 10.1016/j.cbi.2016.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/16/2015] [Accepted: 02/10/2016] [Indexed: 02/05/2023]
|
19
|
Relationships of human α/β hydrolase fold proteins and other organophosphate-interacting proteins. Chem Biol Interact 2016; 259:343-351. [PMID: 27109753 DOI: 10.1016/j.cbi.2016.04.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
Organophosphates (OPs) are either found in nature or synthetized for use as pesticides, flame retardants, neurotoxic warfare agents or drugs (cholinergic enhancers in Alzheimer's disease and myasthenia gravis, or inhibitors of lipases in metabolic diseases). Because of the central role of acetylcholinesterase cholinergic neurotransmission in humans, one of the main purposes for using OPs is inactivation of the enzyme by phosphorylation of the nucleophilic serine residue in the active center. However, hundreds of serine hydrolases are expressed in the human proteome, and many of them are potential targets for OP adduction. In this review, we first situate the α/β hydrolase fold proteins among the distinctively folded proteins known to interact with OPs, in particular the different lipases, peptidases, and enzymes hydrolyzing OPs. Second, we compile the human α/β hydrolases and review those that have been experimentally shown to interact with OPs. Among the 120 human α/β hydrolase fold proteins, 102 have a serine in the consensus GXSXG pentapeptide compatible with an active site, 6 have an aspartate or a cysteine as the active site nucleophile residue, and 12 evidently lack an active site. 76 of the 120 have been experimentally shown to bind an OP.
Collapse
|
20
|
Tuin AW, Mol MAE, van den Berg RM, Fidder A, van der Marel GA, Overkleeft HS, Noort D. Activity-Based Protein Profiling Reveals Broad Reactivity of the Nerve Agent Sarin. Chem Res Toxicol 2016; 22:683-9. [PMID: 19226147 DOI: 10.1021/tx8004218] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Elucidation of noncholinesterase protein targets of organophosphates, and nerve agents in particular, may reveal additional mechanisms for their high toxicity as well as clues for novel therapeutic approaches toward intoxications with these agents. Within this framework, we here describe the synthesis of the activity-based probe 3, which contains a phosphonofluoridate moiety, a P-Me moiety, and a biotinylated O-alkyl group, and its use in activity-based protein profiling with two relevant biological samples, that is, rhesus monkey liver and cultured human A549 lung cells. In this way, we have unearthed eight serine hydrolases (fatty acid synthase, acylpeptide hydrolase, dipeptidyl peptidase 9, prolyl oligopeptidase, carboxylesterase, long-chain acyl coenzyme A thioesterase, PAF acetylhydrolase 1b, and esterase D/S-formyl glutathione hydrolase) as targets that are modified by the nerve agent sarin. It is also shown that the newly developed probe 3 might find its way into the development of alternative, less laborious purification protocols for human butyrylcholinesterase, a potent bioscavenger currently under clinical investigation as a prophylactic/therapeutic for nerve agent intoxications.
Collapse
Affiliation(s)
- Adriaan W Tuin
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Business Unit Biological and Chemical Protection, TNO Defense, Security and Safety, P.O. Box 45, 2280 AA Rijswijk, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
21
|
Carboxylesterases: General detoxifying enzymes. Chem Biol Interact 2016; 259:327-331. [PMID: 26892220 DOI: 10.1016/j.cbi.2016.02.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/15/2016] [Accepted: 02/11/2016] [Indexed: 11/22/2022]
Abstract
Carboxylesterases (CE) are members of the esterase family of enzymes, and as their name suggests, they are responsible for the hydrolysis of carboxylesters into the corresponding alcohol and carboxylic acid. To date, no endogenous CE substrates have been identified and as such, these proteins are thought to act as a mechanism to detoxify ester-containing xenobiotics. As a consequence, they are expressed in tissues that might be exposed to such agents (lung and gut epithelia, liver, kidney, etc.). CEs demonstrate very broad substrate specificities and can hydrolyze compounds as diverse as cocaine, oseltamivir (Tamiflu), permethrin and irinotecan. In addition, these enzymes are irreversibly inhibited by organophosphates such as Sarin and Tabun. In this overview, we will compare and contrast the two human enzymes that have been characterized, and evaluate the biology of the interaction of these proteins with organophosphates (principally nerve agents).
Collapse
|
22
|
Arena de Souza V, Scott DJ, Nettleship JE, Rahman N, Charlton MH, Walsh MA, Owens RJ. Comparison of the Structure and Activity of Glycosylated and Aglycosylated Human Carboxylesterase 1. PLoS One 2015; 10:e0143919. [PMID: 26657071 PMCID: PMC4676782 DOI: 10.1371/journal.pone.0143919] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/11/2015] [Indexed: 11/25/2022] Open
Abstract
Human Carboxylesterase 1 (hCES1) is the key liver microsomal enzyme responsible for detoxification and metabolism of a variety of clinical drugs. To analyse the role of the single N-linked glycan on the structure and activity of the enzyme, authentically glycosylated and aglycosylated hCES1, generated by mutating asparagine 79 to glutamine, were produced in human embryonic kidney cells. Purified enzymes were shown to be predominantly trimeric in solution by analytical ultracentrifugation. The purified aglycosylated enzyme was found to be more active than glycosylated hCES1 and analysis of enzyme kinetics revealed that both enzymes exhibit positive cooperativity. Crystal structures of hCES1 a catalytically inactive mutant (S221A) and the aglycosylated enzyme were determined in the absence of any ligand or substrate to high resolutions (1.86 Å, 1.48 Å and 2.01 Å, respectively). Superposition of all three structures showed only minor conformational differences with a root mean square deviations of around 0.5 Å over all Cα positions. Comparison of the active sites of these un-liganded enzymes with the structures of hCES1-ligand complexes showed that side-chains of the catalytic triad were pre-disposed for substrate binding. Overall the results indicate that preventing N-glycosylation of hCES1 does not significantly affect the structure or activity of the enzyme.
Collapse
Affiliation(s)
- Victoria Arena de Souza
- UK OPPF-UK, The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- Division of Structural Biology, Henry Wellcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - David J. Scott
- The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, Leicestershire, United Kingdom
| | - Joanne E. Nettleship
- UK OPPF-UK, The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- Division of Structural Biology, Henry Wellcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Nahid Rahman
- UK OPPF-UK, The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- Division of Structural Biology, Henry Wellcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Michael H. Charlton
- Chroma Therapeutics Ltd., 93 Innovation Drive Milton Park, Abingdon, United Kingdom
| | - Martin A. Walsh
- The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
- * E-mail: (MAW); (RJO)
| | - Raymond J. Owens
- UK OPPF-UK, The Research Complex at Harwell, Rutherford Appleton Laboratory Harwell Oxford, Oxfordshire, United Kingdom
- Division of Structural Biology, Henry Wellcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
- * E-mail: (MAW); (RJO)
| |
Collapse
|
23
|
Sun DX, Ge GB, Dong PP, Cao YF, Fu ZW, Ran RX, Wu X, Zhang YY, Hua HM, Zhao Z, Fang ZZ. Inhibition behavior of fructus psoraleae’s ingredients towards human carboxylesterase 1 (hCES1). Xenobiotica 2015; 46:503-10. [DOI: 10.3109/00498254.2015.1091521] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
24
|
Kirby SD, Norris J, Sweeney R, Bahnson BJ, Cerasoli DM. A rationally designed mutant of plasma platelet-activating factor acetylhydrolase hydrolyzes the organophosphorus nerve agent soman. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1809-1815. [PMID: 26343853 DOI: 10.1016/j.bbapap.2015.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/21/2015] [Accepted: 09/01/2015] [Indexed: 01/20/2023]
Abstract
Organophosphorus compounds (OPs) such as sarin and soman are some of the most toxic chemicals synthesized by man. They exert toxic effects by inactivating acetylcholinesterase (AChE) and bind secondary target protein. Organophosphorus compounds are hemi-substrates for enzymes of the serine hydrolase superfamily. Enzymes can be engineered by amino acid substitution into OP-hydrolyzing variants (bioscavengers) and used as therapeutics. Some enzymes associated with lipoproteins, such as human plasma platelet-activating factor acetylhydrolase (pPAF-AH), are also inhibited by OPs; these proteins have largely been ignored for engineering purposes because of complex interfacial kinetics and a lack of structural data. We have expressed active human pPAF-AH in bacteria and previously solved the crystal structure of this enzyme with OP adducts. Using these structures as a guide, we created histidine mutations near the active site of pPAF-AH (F322H, W298H, L153H) in an attempt to generate novel OP-hydrolase activity. Wild-type pPAF-AH, L153H, and F322H have essentially no hydrolytic activity against the nerve agents tested. In contrast, the W298H mutant displayed novel somanase activity with a kcat of 5min(-1) and a KM of 590μM at pH7.5. There was no selective preference for hydrolysis of any of the four soman stereoisomers.
Collapse
Affiliation(s)
- Stephen D Kirby
- U.S. Army Medical Research Institute of Chemical Defense, APG, MD 21010, United States; Department of Chemistry & Biochemistry, University of Delaware, Newark, DE 19716, United States.
| | - Joseph Norris
- U.S. Army Medical Research Institute of Chemical Defense, APG, MD 21010, United States
| | - Richard Sweeney
- U.S. Army Medical Research Institute of Chemical Defense, APG, MD 21010, United States
| | - Brian J Bahnson
- Department of Chemistry & Biochemistry, University of Delaware, Newark, DE 19716, United States
| | - Douglas M Cerasoli
- U.S. Army Medical Research Institute of Chemical Defense, APG, MD 21010, United States
| |
Collapse
|
25
|
Palermo G, Campomanes P, Cavalli A, Rothlisberger U, De Vivo M. Anandamide Hydrolysis in FAAH Reveals a Dual Strategy for Efficient Enzyme-Assisted Amide Bond Cleavage via Nitrogen Inversion. J Phys Chem B 2014; 119:789-801. [DOI: 10.1021/jp5052276] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Giulia Palermo
- Department
of Drug Discovery and Development, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Pablo Campomanes
- Laboratory
of Computational Chemistry and Biochemistry, Institute of Chemical
Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne
(EPFL), CH-1015 Lausanne, Switzerland
| | - Andrea Cavalli
- Department
of Drug Discovery and Development, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, I-40126 Bologna, Italy
| | - Ursula Rothlisberger
- Laboratory
of Computational Chemistry and Biochemistry, Institute of Chemical
Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne
(EPFL), CH-1015 Lausanne, Switzerland
| | - Marco De Vivo
- Department
of Drug Discovery and Development, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
26
|
Legler PM, Boisvert SM, Compton JR, Millard CB. Development of organophosphate hydrolase activity in a bacterial homolog of human cholinesterase. Front Chem 2014; 2:46. [PMID: 25077141 PMCID: PMC4100338 DOI: 10.3389/fchem.2014.00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/19/2014] [Indexed: 01/10/2023] Open
Abstract
We applied a combination of rational design and directed evolution (DE) to Bacillus subtilis p-nitrobenzyl esterase (pNBE) with the goal of enhancing organophosphorus acid anhydride hydrolase (OPAAH) activity. DE started with a designed variant, pNBE A107H, carrying a histidine homologous with human butyrylcholinesterase G117H to find complementary mutations that further enhance its OPAAH activity. Five sites were selected (G105, G106, A107, A190, and A400) within a 6.7 Å radius of the nucleophilic serine Oγ. All 95 variants were screened for esterase activity with a set of five substrates: pNP-acetate, pNP-butyrate, acetylthiocholine, butyrylthiocholine, or benzoylthiocholine. A microscale assay for OPAAH activity was developed for screening DE libraries. Reductions in esterase activity were generally concomitant with enhancements in OPAAH activity. One variant, A107K, showed an unexpected 7-fold increase in its k cat/K m for benzoylthiocholine, demonstrating that it is also possible to enhance the cholinesterase activity of pNBE. Moreover, DE resulted in at least three variants with modestly enhanced OPAAH activity compared to wild type pNBE. A107H/A190C showed a 50-fold increase in paraoxonase activity and underwent a slow time- and temperature-dependent change affecting the hydrolysis of OPAA and ester substrates. Structural analysis suggests that pNBE may represent a precursor leading to human cholinesterase and carboxylesterase 1 through extension of two vestigial specificity loops; a preliminary attempt to transfer the Ω-loop of BChE into pNBE is described. Unlike butyrylcholinesterase and pNBE, introducing a G143H mutation (equivalent to G117H) did not confer detectable OP hydrolase activity on human carboxylesterase 1 (hCE1). We discuss the use of pNBE as a surrogate scaffold for the mammalian esterases, and the importance of the oxyanion-hole residues for enhancing the OPAAH activity of selected serine hydrolases.
Collapse
Affiliation(s)
- Patricia M. Legler
- Naval Research Laboratory, Center for Bio/Molecular Science and EngineeringWashington, DC, USA
| | | | | | - Charles B. Millard
- United States Army Medical Research and Materiel CommandFort Detrick, MD, USA
| |
Collapse
|
27
|
Reiter G, Müller S, Koller M, Thiermann H, Worek F. In vitro toxicokinetic studies of cyclosarin: Molecular mechanisms of elimination. Toxicol Lett 2014; 227:1-11. [DOI: 10.1016/j.toxlet.2014.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 11/17/2022]
|
28
|
Jackson CJ, Liu JW, Carr PD, Younus F, Coppin C, Meirelles T, Lethier M, Pandey G, Ollis DL, Russell RJ, Weik M, Oakeshott JG. Structure and function of an insect α-carboxylesterase (αEsterase7) associated with insecticide resistance. Proc Natl Acad Sci U S A 2013; 110:10177-82. [PMID: 23733941 PMCID: PMC3690851 DOI: 10.1073/pnas.1304097110] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Insect carboxylesterases from the αEsterase gene cluster, such as αE7 (also known as E3) from the Australian sheep blowfly Lucilia cuprina (LcαE7), play an important physiological role in lipid metabolism and are implicated in the detoxification of organophosphate (OP) insecticides. Despite the importance of OPs to agriculture and the spread of insect-borne diseases, the molecular basis for the ability of α-carboxylesterases to confer OP resistance to insects is poorly understood. In this work, we used laboratory evolution to increase the thermal stability of LcαE7, allowing its overexpression in Escherichia coli and structure determination. The crystal structure reveals a canonical α/β-hydrolase fold that is very similar to the primary target of OPs (acetylcholinesterase) and a unique N-terminal α-helix that serves as a membrane anchor. Soaking of LcαE7 crystals in OPs led to the capture of a crystallographic snapshot of LcαE7 in its phosphorylated state, which allowed comparison with acetylcholinesterase and rationalization of its ability to protect insects against the effects of OPs. Finally, inspection of the active site of LcαE7 reveals an asymmetric and hydrophobic substrate binding cavity that is well-suited to fatty acid methyl esters, which are hydrolyzed by the enzyme with specificity constants (∼10(6) M(-1) s(-1)) indicative of a natural substrate.
Collapse
Affiliation(s)
- Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT 0200, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Muthukrishnan S, Shete VS, Sanan TT, Vyas S, Oottikkal S, Porter LM, Magliery TJ, Hadad CM. Mechanistic Insights into the Hydrolysis of Organophosphorus Compounds by Paraoxonase-1: Exploring the Limits of Substrate Tolerance in a Promiscuous Enzyme. J PHYS ORG CHEM 2012; 25:1247-1260. [PMID: 23946555 PMCID: PMC3740977 DOI: 10.1002/poc.3002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We designed, synthesized and screened a library of analogs of the organophosphate pesticide metabolite paraoxon against a recombinant variant of human serum paraoxonase-1. Alterations of both the aryloxy leaving group and the retained alkyl chains of paraoxon analogs resulted in substantial changes to binding and hydrolysis, as measured directly by spectrophotometric methods or in competition experiments with paraoxon. Increases or decreases in the steric bulk of the retained groups generally reduced the rate of hydrolysis, while modifications of the leaving group modulated both binding and turnover. Studies on the hydrolysis of phosphoryl azide analogs as well as amino-modified paraoxon analogs, the former being developed as photo-affinity labels, found enhanced tolerance of structural modifications, when compared with O-alkyl substituted molecules. Results from computational modeling predict a predominant active site binding mode for these molecules which is consistent with several proposed catalytic mechanisms in the literature, and from which a molecular-level explanation of the experimental trends is attempted. Overall, the results of this study suggest that while paraoxonase-1 is a promiscuous enzyme, there are substantial constraints in the active site pocket, which may relate to both the leaving group and the retained portion of paraoxon analogs.
Collapse
Affiliation(s)
| | - Vivekanand S. Shete
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Toby. T. Sanan
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Shubham Vyas
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Shameema Oottikkal
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Lauren M. Porter
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Thomas J. Magliery
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
- Department of Biochemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| | - Christopher M. Hadad
- Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio, 43210, U.S.A
| |
Collapse
|
30
|
Greenblatt HM, Otto TC, Kirkpatrick MG, Kovaleva E, Brown S, Buchman G, Cerasoli DM, Sussman JL. Structure of recombinant human carboxylesterase 1 isolated from whole cabbage looper larvae. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:269-72. [PMID: 22442219 PMCID: PMC3310527 DOI: 10.1107/s1744309112003326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 01/25/2012] [Indexed: 11/11/2022]
Abstract
The use of whole insect larvae as a source of recombinant proteins offers a more cost-effective method of producing large quantities of human proteins than conventional cell-culture approaches. Human carboxylesterase 1 has been produced in and isolated from whole Trichoplusia ni larvae. The recombinant protein was crystallized and its structure was solved to 2.2 resolution. The results indicate that the larvae-produced enzyme is essentially identical to that isolated from cultured Sf21 cells, supporting the use of this expression system to produce recombinant enzymes for crystallization studies.
Collapse
Affiliation(s)
- Harry M. Greenblatt
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tamara C. Otto
- Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Melanie G. Kirkpatrick
- Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Elena Kovaleva
- Chesapeake PERL Inc., 8510A Corridor Road, Savage, MD 20763, USA
| | - Susan Brown
- Chesapeake PERL Inc., 8510A Corridor Road, Savage, MD 20763, USA
| | - George Buchman
- Chesapeake PERL Inc., 8510A Corridor Road, Savage, MD 20763, USA
| | - Douglas M. Cerasoli
- Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Joel L. Sussman
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
31
|
Edwards JS, Kumbhar A, Roberts A, Hemmert AC, Edwards CC, Potter PM, Redinbo MR. Immobilization of active human carboxylesterase 1 in biomimetic silica nanoparticles. Biotechnol Prog 2011; 27:863-9. [PMID: 21509954 DOI: 10.1002/btpr.604] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/26/2011] [Indexed: 11/12/2022]
Abstract
The encapsulation of proteins in biomimetic silica has recently been shown to successfully maintain enzymes in their active state. Organophosphate (OP) compounds are used as pesticides as well as potent chemical warfare nerve agents. Because these toxicants are life threatening, we sought to generate biomimetic silicas capable of responding to OPs. Here, we present the silica encapsulation of human drug metabolism enzyme carboxylesterase 1 (hCE1) in the presence of a range of catalysts. hCE1 was successfully encapsulated into silica particles when lysozyme or the peptide R5 were used as catalysts; in contrast, polyethyleneimine, a catalyst used to encapuslate other enzymes, did not facilitate hCE1 entrapment. hCE1 silica particles in a column chromatography format respond to the presence of the OP pesticides paraoxon and dimethyl-p-nitrophenyl phosphate in solution. These results may lead to novel approaches to detect OP pesticides or other weaponized agents that bind hCE1.
Collapse
Affiliation(s)
- Jonathan S Edwards
- Dept. of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Hemmert AC, Otto TC, Chica RA, Wierdl M, Edwards JS, Lewis SL, Edwards CC, Tsurkan L, Cadieux CL, Kasten SA, Cashman JR, Mayo SL, Potter PM, Cerasoli DM, Redinbo MR. Nerve agent hydrolysis activity designed into a human drug metabolism enzyme. PLoS One 2011; 6:e17441. [PMID: 21445272 PMCID: PMC3060870 DOI: 10.1371/journal.pone.0017441] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 02/02/2011] [Indexed: 12/04/2022] Open
Abstract
Organophosphorus (OP) nerve agents are potent suicide inhibitors of the essential neurotransmitter-regulating enzyme acetylcholinesterase. Due to their acute toxicity, there is significant interest in developing effective countermeasures to OP poisoning. Here we impart nerve agent hydrolysis activity into the human drug metabolism enzyme carboxylesterase 1. Using crystal structures of the target enzyme in complex with nerve agent as a guide, a pair of histidine and glutamic acid residues were designed proximal to the enzyme's native catalytic triad. The resultant variant protein demonstrated significantly increased rates of reactivation following exposure to sarin, soman, and cyclosarin. Importantly, the addition of these residues did not alter the high affinity binding of nerve agents to this protein. Thus, using two amino acid substitutions, a novel enzyme was created that efficiently converted a group of hemisubstrates, compounds that can start but not complete a reaction cycle, into bona fide substrates. Such approaches may lead to novel countermeasures for nerve agent poisoning.
Collapse
Affiliation(s)
- Andrew C. Hemmert
- Department of Biochemistry/Biophysics and Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Tamara C. Otto
- United States Army Medical Research Institute for Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Roberto A. Chica
- Department of Biology and Chemistry, California Institute of Technology, Pasadena, California, United States of America
| | - Monika Wierdl
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Jonathan S. Edwards
- Department of Biochemistry/Biophysics and Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Steven L. Lewis
- Department of Biochemistry/Biophysics and Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Carol C. Edwards
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Lyudmila Tsurkan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - C. Linn Cadieux
- United States Army Medical Research Institute for Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Shane A. Kasten
- United States Army Medical Research Institute for Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - John R. Cashman
- Human BioMolecular Research Institute, San Diego, California, United States of America
| | - Stephen L. Mayo
- Department of Biology and Chemistry, California Institute of Technology, Pasadena, California, United States of America
| | - Philip M. Potter
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Douglas M. Cerasoli
- United States Army Medical Research Institute for Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Matthew R. Redinbo
- Department of Biochemistry/Biophysics and Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
33
|
Structural approach to the aging of phosphylated cholinesterases. Chem Biol Interact 2010; 187:157-62. [DOI: 10.1016/j.cbi.2010.03.027] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Revised: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 12/18/2022]
|
34
|
Sanan TT, Muthukrishnan S, Beck JM, Tao P, Hayes CJ, Otto TC, Cerasoli DM, Lenz DE, Hadad CM. Computational Modeling of Human Paraoxonase 1: Preparation of Protein Models, Binding Studies, and Mechanistic Insights. J PHYS ORG CHEM 2010; 23:357-369. [PMID: 24077808 PMCID: PMC3783361 DOI: 10.1002/poc.1678] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The enzyme human paraoxonase 1 (huPON1) has demonstrated significant potential for use as a bioscavenger for treatment of exposure to organophosphorus (OP) nerve agents. Herein we report the development of protein models for the human isoform derived from a crystal structure of a chimeric version of the protein (pdb ID: 1V04) and a homology model derived from the related enzyme diisopropylfluorophosphatase (pdb ID: 1XHR). From these structural models, binding modes for OP substrates are predicted, and these poses are found to orient substrates in proximity to residues known to modulate specificity of the enzyme. Predictions are made with regard to the role that residues play in altering substrate binding and turnover, in particular with regard to the stereoselectivity of the enzyme, and the known differences in activity related to a natural polymorphism in the enzyme. Potential mechanisms of action of the protein for catalytic hydrolysis of OP substrates are also evaluated in light of the proposed binding modes.
Collapse
Affiliation(s)
- Toby T. Sanan
- Department of Chemistry, 100 West 18 Avenue, Ohio State University, Columbus, Ohio 43210
| | | | - Jeremy M. Beck
- Department of Chemistry, 100 West 18 Avenue, Ohio State University, Columbus, Ohio 43210
| | - Peng Tao
- Department of Chemistry, 100 West 18 Avenue, Ohio State University, Columbus, Ohio 43210
| | - Carrigan J. Hayes
- Department of Chemistry, 100 West 18 Avenue, Ohio State University, Columbus, Ohio 43210
| | - Tamara C. Otto
- 3100 Ricketts Point Rd, Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland 21010
| | - Douglas M. Cerasoli
- 3100 Ricketts Point Rd, Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland 21010
| | - David E. Lenz
- 3100 Ricketts Point Rd, Physiology and Immunology Branch, Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland 21010
| | - Christopher M. Hadad
- Department of Chemistry, 100 West 18 Avenue, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
35
|
Hemmert AC, Otto TC, Wierdl M, Edwards CC, Fleming CD, MacDonald M, Cashman JR, Potter PM, Cerasoli DM, Redinbo MR. Human carboxylesterase 1 stereoselectively binds the nerve agent cyclosarin and spontaneously hydrolyzes the nerve agent sarin. Mol Pharmacol 2010; 77:508-16. [PMID: 20051531 DOI: 10.1124/mol.109.062356] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Organophosphorus (OP) nerve agents are potent toxins that inhibit cholinesterases and produce a rapid and lethal cholinergic crisis. Development of protein-based therapeutics is being pursued with the goal of preventing nerve agent toxicity and protecting against the long-term side effects of these agents. The drug-metabolizing enzyme human carboxylesterase 1 (hCE1) is a candidate protein-based therapeutic because of its similarity in structure and function to the cholinesterase targets of nerve agent poisoning. However, the ability of wild-type hCE1 to process the G-type nerve agents sarin and cyclosarin has not been determined. We report the crystal structure of hCE1 in complex with the nerve agent cyclosarin. We further use stereoselective nerve agent analogs to establish that hCE1 exhibits a 1700- and 2900-fold preference for the P(R) enantiomers of analogs of soman and cyclosarin, respectively, and a 5-fold preference for the P(S) isomer of a sarin analog. Finally, we show that for enzyme inhibited by racemic mixtures of bona fide nerve agents, hCE1 spontaneously reactivates in the presence of sarin but not soman or cyclosarin. The addition of the neutral oxime 2,3-butanedione monoxime increases the rate of reactivation of hCE1 from sarin inhibition by more than 60-fold but has no effect on reactivation with the other agents examined. Taken together, these data demonstrate that hCE1 is only reactivated after inhibition with the more toxic P(S) isomer of sarin. These results provide important insights toward the long-term goal of designing novel forms of hCE1 to act as protein-based therapeutics for nerve agent detoxification.
Collapse
Affiliation(s)
- Andrew C Hemmert
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sanson B, Nachon F, Colletier JP, Froment MT, Toker L, Greenblatt HM, Sussman JL, Ashani Y, Masson P, Silman I, Weik M. Crystallographic snapshots of nonaged and aged conjugates of soman with acetylcholinesterase, and of a ternary complex of the aged conjugate with pralidoxime. J Med Chem 2009; 52:7593-603. [PMID: 19642642 DOI: 10.1021/jm900433t] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Organophosphate compounds (OP) are potent inhibitors of acetylcholinesterases (AChEs) and can cause lethal poisoning in humans. Inhibition of AChEs by the OP soman involves phosphonylation of the catalytic serine, and subsequent dealkylation produces a form known as the "aged" enzyme. The nonaged form can be reactivated to a certain extent by nucleophiles, such as pralidoxime (2-PAM), whereas aged forms of OP-inhibited AChEs are totally resistant to reactivation. Here, we solved the X-ray crystal structures of AChE from Torpedo californica (TcAChE) conjugated with soman before and after aging. The absolute configuration of the soman stereoisomer adduct in the nonaged conjugate is P(S)C(R). A structural reorientation of the catalytic His440 side chain was observed during the aging process. Furthermore, the crystal structure of the ternary complex of the aged conjugate with 2-PAM revealed that the orientation of the oxime function does not permit nucleophilic attack on the phosphorus atom, thus providing a plausible explanation for its failure to reactivate the aged soman/AChE conjugate. Together, these three crystal structures provide an experimental basis for the design of new reactivators.
Collapse
Affiliation(s)
- Benoît Sanson
- Laboratoire de Biophysique Moléculaire, Institut de Biologie Structurale Jean-Pierre Ebel, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Université Joseph Fourier, 41 Rue Jules Horowitz, 38027 Grenoble, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Butyrylcholinesterase for protection from organophosphorus poisons: catalytic complexities and hysteretic behavior. Arch Biochem Biophys 2009; 494:107-20. [PMID: 20004171 DOI: 10.1016/j.abb.2009.12.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 11/24/2009] [Accepted: 12/01/2009] [Indexed: 12/13/2022]
Abstract
Butyrylcholinesterase is a promiscuous enzyme that displays complex kinetic behavior. It is toxicologically important because it detoxifies organophosphorus poisons (OP) by making a covalent bond with the OP. The OP and the butyrylcholinesterase are both inactivated in the process. Inactivation of butyrylcholinesterase has no adverse effects. However, inactivation of acetylcholinesterase in nerve synapses can be lethal. OP-inhibited butyrylcholinesterase and acetylcholinesterase can be reactivated with oximes provided the OP has not aged. Strategies for preventing the toxicity of OP include (a) treatment with an OP scavenger, (b) reaction of non-aged enzyme with oximes, (c) reactivation of aged enzyme, (d) slowing down aging with peripheral site ligands, and (e) design of mutants that rapidly hydrolyze OP. Option (a) has progressed through phase I clinical trials with human butyrylcholinesterase. Option (b) is in routine clinical use. The others are at the basic research level. Butyrylcholinesterase displays complex kinetic behavior including activation by positively charged esters, ability to hydrolyze amides, and a lag time (hysteresis) preceding hydrolysis of benzoylcholine and N-methylindoxyl acetate. Mass spectrometry has identified new OP binding motifs on tyrosine and lysine in proteins that have no active site serine. It is proposed, but not yet proven, that low dose exposure involves OP modification of proteins that have no active site serine.
Collapse
|
38
|
Makhaeva GF, Aksinenko AY, Sokolov VB, Serebryakova OG, Richardson RJ. Synthesis of organophosphates with fluorine-containing leaving groups as serine esterase inhibitors with potential for Alzheimer disease therapeutics. Bioorg Med Chem Lett 2009; 19:5528-30. [PMID: 19717305 DOI: 10.1016/j.bmcl.2009.08.065] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Revised: 08/14/2009] [Accepted: 08/18/2009] [Indexed: 11/27/2022]
Abstract
Acetylcholinesterase and butyrylcholinesterase inhibitors are potential cognition enhancers in Alzheimer disease. O,O-Dialkylphosphate inhibitors with 1-substituted 2,2,2-trifluoroethoxy leaving groups were synthesized by phosphonate-phosphate rearrangement. Substituents in the 1-position of the leaving group along with the O-alkyl groups modulated potency and selectivity against acetylcholinesterase, butyrylcholinesterase, and carboxylesterase.
Collapse
Affiliation(s)
- Galina F Makhaeva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | | | | | | | | |
Collapse
|
39
|
Lamango NS, Duverna R, Zhang W, Ablordeppey SY. Porcine Liver Carboxylesterase Requires Polyisoprenylation for High Affinity Binding to Cysteinyl Substrates. ACTA ACUST UNITED AC 2009; 2:12-27. [PMID: 20664805 DOI: 10.2174/1874940200902010012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The polyisoprenylation pathway enzymes have been the focus of numerous studies to better understand the roles of polyisoprenylated proteins in eukaryotic cells and to identify novel targets against diseases such as cancer. The final step of the pathway is a reversible reaction catalyzed by isoprenyl carboxylmethyl transferase (icmt) whose products are then hydrolyzed by polyisoprenylated methylated protein methyl esterase (PMPMEase). Unlike the other pathway enzymes, the esterase has received little attention. We recently purified PMPMEase from porcine liver using an S-polyisoprenylated cysteine methyl ester substrate-dependent screening assay. However, no data is available showing its relative interaction with structurally diverse substrates. As such, its role as the putative endogenous PMPMEase has not been demonstrated. A series of substrates with S-alkyl substituents ranging from 2 to 20 carbons, including the two moieties found in polyisoprenylated proteins, were synthesized. Enzyme kinetics analysis revealed a 33-fold increase in affinity (K(M) values) from ethyl- (C-2, 505+/-63 microM), prenyl- (C-5, 294+/-25 microM), trans-geranyl- (C-10, 87+/-12 microM), trans, trans-farnesyl- (C-15, 29+/-2.2 microM) to all trans-geranylgeranyl- (C-20-, 15+/-2.7 microM) based analogs. Comparative molecular field analysis of the data yielded a cross-validated q(2) of 0.863+/-0.365 and a final R(2) of 0.995. Since the substrates with the S-trans, trans-farnesyl and S-all trans-geranylgeranyl moieties that occur in proteins show the highest affinity towards PMPMEase and are not hydrolyzed by the cholinesterases, the results suggest that polyisoprenylated proteins are the endogenous substrates of this esterase. The results suggest design strategies for high affinity and selective inhibitors of PMPMEase.
Collapse
Affiliation(s)
- Nazarius S Lamango
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | | | | | | |
Collapse
|
40
|
Epstein TM, Samanta U, Kirby SD, Cerasoli DM, Bahnson BJ. Crystal structures of brain group-VIII phospholipase A2 in nonaged complexes with the organophosphorus nerve agents soman and sarin. Biochemistry 2009; 48:3425-35. [PMID: 19271773 DOI: 10.1021/bi8023527] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Insecticide and nerve agent organophosphorus (OP) compounds are potent inhibitors of the serine hydrolase superfamily of enzymes. Nerve agents, such as sarin, soman, tabun, and VX exert their toxicity by inhibiting human acetycholinesterase at nerve synapses. Following the initial phosphonylation of the active site serine, the enzyme may reactivate spontaneously or through reaction with an appropriate nucleophilic oxime. Alternatively, the enzyme-nerve agent complex can undergo a secondary process, called "aging", which dealkylates the nerve agent adduct and results in a product that is highly resistant to reactivation by any known means. Here we report the structures of paraoxon, soman, and sarin complexes of group-VIII phospholipase A2 from bovine brain. In each case, the crystal structures indicate a nonaged adduct; a stereoselective preference for binding of the P(S)C(S) isomer of soman and the P(S) isomer of sarin was also noted. The stability of the nonaged complexes was corroborated by trypsin digest and electrospray ionization mass spectrometry, which indicates nonaged complexes are formed with diisopropylfluorophosphate, soman, and sarin. The P(S) stereoselectivity for reaction with sarin was confirmed by reaction of racemic sarin, followed by gas chromatography/mass spectrometry using a chiral column to separate and quantitate each stereoisomer. The P(S) stereoisomers of soman and sarin are known to be the more toxic stereoisomers, as they react preferentially to inhibit human acetylcholinesterase. The results obtained for nonaged complexes of group-VIII phospholipase A2 are compared to those obtained for other serine hydrolases and discussed to partly explain determinants of OP aging. Furthermore, structural insights can now be exploited to engineer variant versions of this enzyme with enhanced nerve agent binding and hydrolysis functions.
Collapse
Affiliation(s)
- Todd M Epstein
- Department of Chemistry & Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | | | | | | | | |
Collapse
|
41
|
Samanta U, Kirby SD, Srinivasan P, Cerasoli DM, Bahnson BJ. Crystal structures of human group-VIIA phospholipase A2 inhibited by organophosphorus nerve agents exhibit non-aged complexes. Biochem Pharmacol 2009; 78:420-9. [PMID: 19394314 DOI: 10.1016/j.bcp.2009.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/16/2009] [Accepted: 04/16/2009] [Indexed: 11/17/2022]
Abstract
The enzyme group-VIIA phospholipase A2 (gVIIA-PLA2) is bound to lipoproteins in human blood and hydrolyzes the ester bond at the sn-2 position of phospholipid substrates with a short sn-2 chain. The enzyme belongs to a serine hydrolase superfamily of enzymes, which react with organophosphorus (OP) nerve agents. OPs ultimately exert their toxicity by inhibiting human acetycholinesterase at nerve synapses, but may additionally have detrimental effects through inhibition of other serine hydrolases. We have solved the crystal structures of gVIIA-PLA2 following inhibition with the OPs diisopropylfluorophosphate, sarin, soman and tabun. The sarin and soman complexes displayed a racemic mix of P(R) and P(S) stereoisomers at the P-chiral center. The tabun complex displayed only the P(R) stereoisomer in the crystal. In all cases, the crystal structures contained intact OP adducts that had not aged. Aging refers to a secondary process OP complexes can go through, which dealkylates the nerve agent adduct and results in a form that is highly resistant to either spontaneous or oxime-mediated reactivation. Non-aged OP complexes of the enzyme were corroborated by trypsin digest and matrix-assisted laser desorption ionization mass spectrometry of OP-enzyme complexes. The lack of stereoselectivity of sarin reaction was confirmed by gas chromatography/mass spectrometry using a chiral column to separate and quantitate the unbound stereoisomers of sarin following incubation with enzyme. The structural details and characterization of nascent reactivity of several toxic nerve agents is discussed with a long-term goal of developing gVIIA-PLA2 as a catalytic bioscavenger of OP nerve agents.
Collapse
Affiliation(s)
- Uttamkumar Samanta
- Department of Chemistry & Biochemistry, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | |
Collapse
|
42
|
Grigoryan H, Li B, Anderson EK, Xue W, Nachon F, Lockridge O, Schopfer LM. Covalent binding of the organophosphorus agent FP-biotin to tyrosine in eight proteins that have no active site serine. Chem Biol Interact 2009; 180:492-8. [PMID: 19539807 DOI: 10.1016/j.cbi.2009.03.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Revised: 03/23/2009] [Accepted: 03/24/2009] [Indexed: 11/25/2022]
Abstract
Organophosphorus (OP) esters are known to bind covalently to the active site serine of enzymes in the serine hydrolase family. It was a surprise to find that proteins with no active site serine are also covalently modified by OP. The binding site in albumin, transferrin, and tubulin was identified as tyrosine. The goal of the present work was to determine whether binding to tyrosine is a general phenomenon. Fourteen proteins were treated with a biotin-tagged organophosphorus agent called FP-biotin. The proteins were digested with trypsin and the labeled peptides enriched by binding to monomeric avidin. Peptides were purified by HPLC and fragmented by collision induced dissociation in a tandem ion trap mass spectrometer. Eight proteins were labeled and six were not. Tyrosine was labeled in human alpha-2-glycoprotein 1 zinc-binding protein (Tyr 138, Tyr 174 and Tyr 181), human kinesin 3C motor domain (Tyr 145), human keratin 1 (Tyr 230), bovine actin (Tyr 55 and Tyr 200), murine ATP synthase beta (Tyr 431), murine adenine nucleotide translocase 1 (Tyr 81), bovine chymotrypsinogen (Tyr 201) and porcine pepsin (Tyr 310). Only 1-3 tyrosines per protein were modified, suggesting that the reactive tyrosine was activated by nearby residues that facilitated ionization of the hydroxyl group of tyrosine. These results suggest that OP binding to tyrosine is a general phenomenon. It is concluded that organophosphorus-reactive proteins include not only enzymes in the serine hydrolase family, but also proteins that have no active site serine. The recognition of a new OP-binding motif to tyrosine suggests new directions to search for mechanisms of long-term effects of OP exposure. Another application is in the search for biomarkers of organophosphorus agent exposure. Previous searches have been limited to serine hydrolases. Now proteins such as albumin and keratin can be considered.
Collapse
Affiliation(s)
- Hasmik Grigoryan
- Eppley Institute, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, United States.
| | | | | | | | | | | | | |
Collapse
|
43
|
Masson P, Rochu D. Catalytic bioscavengers against toxic esters, an alternative approach for prophylaxis and treatments of poisonings. Acta Naturae 2009; 1:68-79. [PMID: 22649587 PMCID: PMC3347506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Bioscavengers are biopharmaceuticals that specifically react with toxicants. Thus, enzymes reacting with poisonous esters can be used as bioscavengers for neutralization of toxic molecules before they reach physiological targets. Parenteral administration of bioscavengers is, therefore, intended for prophylaxis or pre-treatments, emergency and post-exposure treatments of intoxications. These enzymes can also be used for application on skin, mucosa and wounds as active components of topical skin protectants and decontamination solutions. Human butyrylcholinesterase is the first stoichiometric bioscavenger for safe and efficient prophylaxis of organophosphate poisoning. However, huge amounts of a costly enzyme are needed for protection. Thus, the bioscavenger approach will be greatly improved by the use of catalytic bioscavengers. Catalytic bioscavengers are enzymes capable of degrading toxic esters with a turnover. Suitable catalytic bioscavengers are engineered mutants of human enzymes. Efficient mutants of human butyrylcholinesterase have been made that hydrolyze cocaine at a high rate. Mutants of human cholinesterases capable of hydrolyzing OPs have been made, but so far their activity is too low to be of medical interest. Human paraoxonase a promiscuous plasma enzyme is certainly the most promising phosphotriesterase. However, its biotechnology is still in its infancy. Other enzymes and proteins from blood and organs, and secondary biological targets of OPs and carbamates are potential bioscavengers, in particular serum albumin that reacts with OPs and self-reactivates. Lastly, non-human enzymes, phosphotriesterases and oxidases from various bacterial and eukaryotic sources could be used for external use against OP poisoning and for internal use after modifications for immunological compatibility.
Collapse
Affiliation(s)
- Patrick Masson
- Centre de Recherches du Service de Santé des Armées, Toxicology department, La Tronche, France
| | | |
Collapse
|
44
|
Harada T, Nakagawa Y, Wadkins RM, Potter PM, Wheelock CE. Comparison of benzil and trifluoromethyl ketone (TFK)-mediated carboxylesterase inhibition using classical and 3D-quantitative structure-activity relationship analysis. Bioorg Med Chem 2008; 17:149-64. [PMID: 19062296 DOI: 10.1016/j.bmc.2008.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 11/02/2008] [Accepted: 11/06/2008] [Indexed: 10/21/2022]
Abstract
Carboxylesterases are enzymes that hydrolyze a broad suite of endogenous and exogenous ester-containing compounds to the corresponding alcohol and carboxylic acid. These enzymes metabolize a number of therapeutics including the anti-tumor agent CPT-11, the anti-viral drug oseltamivir, and the anti-thrombogenic agent clopidogrel as well as many agrochemicals. In addition, carboxylesterases are involved in lipid homeostasis, including cholesterol metabolism and transport with a proposed role in the development of atherosclerosis. Several different scaffolds capable of inhibiting carboxylesterases have been reported, including organophosphates, carbamates, trifluoromethyl ketone-containing structures (TFKs), and aromatic ethane-1,2-diones. Of these varied groups, only the 1,2-diones evidence carboxylesterase isoform-selectivity, which is an important characteristic for therapeutic application and probing biological mechanisms. This study constructed a series of classical and 3D-QSAR models to examine the physiochemical parameters involved in the observed selectivity of three mammalian carboxylesterases: human intestinal carboxylesterase (hiCE), human carboxylesterase 1 (hCE1), and rabbit carboxylesterase (rCE). CoMFA-based models for the benzil-analogs described 88%, 95% and 76% of observed activity for hiCE, hCE1 and rCE, respectively. For TFK-containing compounds, two distinct models were constructed using either the ketone or gem-diol form of the inhibitor. For all three enzymes, the CoMFA ketone models comprised more biological activity than the corresponding gem-diol models; however the differences were small with described activity for all models ranging from 85-98%. A comprehensive model incorporating both benzil and TFK structures described 92%, 85% and 87% of observed activity for hiCE, hCE1 and rCE, respectively. Both classical and 3D-QSAR analysis showed that the observed isoform-selectivity with the benzil-analogs could be described by the volume parameter. This finding was successfully applied to examine substrate selectivity, demonstrating that the relative volumes of the alcohol and acid moieties of ester-containing substrates were predictive for whether hydrolysis was preferred by hiCE or hCE1. Based upon the integrated benzil and TFK model, the next generation inhibitors should combine the A-ring and the 1,2-dione of the benzil inhibitor with the long alkyl chain of the TFK-inhibitor in order to optimize selectivity and potency. These new inhibitors could be useful for elucidating the role of carboxylesterase activity in fatty acid homeostasis and the development of atherosclerosis as well as effecting the controlled activation of carboxylesterase-based prodrugs in situ.
Collapse
Affiliation(s)
- Toshiyuki Harada
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | | | |
Collapse
|
45
|
Jett DA. Cholinesterase research at the National Institutes of Health, USA. Chem Biol Interact 2008; 175:22-5. [DOI: 10.1016/j.cbi.2008.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 04/03/2008] [Indexed: 10/22/2022]
|
46
|
Masson P, Nachon F, Broomfield CA, Lenz DE, Verdier L, Schopfer LM, Lockridge O. A collaborative endeavor to design cholinesterase-based catalytic scavengers against toxic organophosphorus esters. Chem Biol Interact 2008; 175:273-80. [PMID: 18508040 DOI: 10.1016/j.cbi.2008.04.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 03/18/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
Wild-type human butyrylcholinesterase (BuChE) has proven to be an efficient bioscavenger for protection against nerve agent toxicity. Human acetylcholinesterase (AChE) has a similar potential. A limitation to their usefulness is that both cholinesterases (ChEs) react stoichiometrically with organophosphosphorus (OP) esters. Because OPs can be regarded as pseudo-substrates for which the dephosphylation rate constant is almost zero, several strategies have been attempted to promote the dephosphylation reaction. Oxime-mediated reactivation of phosphylated ChEs generates a turnover, but it is too slow to make pseudo-catalytic scavengers of pharmacological interest. Alternatively, it was hypothesized that ChEs could be converted into OP hydrolases by using rational site-directed mutagenesis based upon the crystal structure of ChEs. The idea was to introduce a nucleophile into the oxyanion hole, at an appropriate position to promote hydrolysis of the phospho-serine bond via a base catalysis mechanism. Such mutants, if they showed the desired catalytic and pharmacokinetic properties, could be used as catalytic scavengers. The first mutant of human BuChE that was capable of hydrolyzing OPs was G117H. It had a slow rate. Crystallographic study of the G117H mutant showed that hydrolysis likely occurs by activation of a water molecule rather than direct nucleophilic attack by H117. Numerous BuChE mutants were made later, but none of them was better than the G117H mutant at hydrolyzing OPs, with the exception of soman. Soman aged too rapidly to be hydrolyzed by G117H. Hydrolysis was however accomplished with the double mutant G117H/E197Q, which did not age after phosphonylation with soman. Multiple mutations in the active center of human and Bungarus AChE led to enzymes displaying low catalytic activity towards OPs and unwanted kinetic complexities. A new generation of human AChE mutants has been designed with the assistance of molecular modelling and computational methods. According to the putative water-activation mechanism of G117H BChE, a new histidine/aspartate dyad was introduced into the active center of human AChE at the optimum location for hydrolysis of the OP adduct. Additional mutations were made for optimizing activity of the new dyad. It is anticipated that these new mutants will have OP hydrolase activity.
Collapse
Affiliation(s)
- Patrick Masson
- Toxicology Department, Enzymology Unit, Centre de Recherches du Service de Santé des Armées, La Tronche Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Holmes RS, Chan J, Cox LA, Murphy WJ, VandeBerg JL. Opossum carboxylesterases: sequences, phylogeny and evidence for CES gene duplication events predating the marsupial-eutherian common ancestor. BMC Evol Biol 2008; 8:54. [PMID: 18289373 PMCID: PMC2266714 DOI: 10.1186/1471-2148-8-54] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Accepted: 02/20/2008] [Indexed: 11/10/2022] Open
Abstract
Background Carboxylesterases (CES) perform diverse metabolic roles in mammalian organisms in the detoxification of a broad range of drugs and xenobiotics and may also serve in specific roles in lipid, cholesterol, pheromone and lung surfactant metabolism. Five CES families have been reported in mammals with human CES1 and CES2 the most extensively studied. Here we describe the genetics, expression and phylogeny of CES isozymes in the opossum and report on the sequences and locations of CES1, CES2 and CES6 'like' genes within two gene clusters on chromosome one. We also discuss the likely sequence of gene duplication events generating multiple CES genes during vertebrate evolution. Results We report a cDNA sequence for an opossum CES and present evidence for CES1 and CES2 like genes expressed in opossum liver and intestine and for distinct gene locations of five opossum CES genes,CES1, CES2.1, CES2.2, CES2.3 and CES6, on chromosome 1. Phylogenetic and sequence alignment studies compared the predicted amino acid sequences for opossum CES with those for human, mouse, chicken, frog, salmon and Drosophila CES gene products. Phylogenetic analyses produced congruent phylogenetic trees depicting a rapid early diversification into at least five distinct CES gene family clusters: CES2, CES1, CES7, CES3, and CES6. Molecular divergence estimates based on a Bayesian relaxed clock approach revealed an origin for the five mammalian CES gene families between 328–378 MYA. Conclusion The deduced amino acid sequence for an opossum cDNA was consistent with its identity as a mammalian CES2 gene product (designated CES2.1). Distinct gene locations for opossum CES1 (1: 446,222,550–446,274,850), three CES2 genes (1: 677,773,395–677,927,030) and a CES6 gene (1: 677,585,520–677,730,419) were observed on chromosome 1. Opossum CES1 and multiple CES2 genes were expressed in liver and intestine. Amino acid sequences for opossum CES1 and three CES2 gene products revealed conserved residues previously reported for human CES1 involved in catalysis, ligand binding, tertiary structure and organelle localization. Phylogenetic studies indicated the gene duplication events which generated ancestral mammalian CES genes predated the common ancestor for marsupial and eutherian mammals, and appear to coincide with the early diversification of tetrapods.
Collapse
Affiliation(s)
- Roger S Holmes
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, TX, USA.
| | | | | | | | | |
Collapse
|
48
|
Wheelock CE, Phillips BM, Anderson BS, Miller JL, Miller MJ, Hammock BD. Applications of carboxylesterase activity in environmental monitoring and toxicity identification evaluations (TIEs). REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2008; 195:117-178. [PMID: 18418956 DOI: 10.1007/978-0-387-77030-7_5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
This review has examined a number of issues surrounding the use of carboxylesterase activity in environmental monitoring. It is clear that carboxylesterases are important enzymes that deserve increased study. This class of enzymes appears to have promise for employment in environmental monitoring with a number of organisms and testing scenarios, and it is appropriate for inclusion in standard monitoring assays. Given the ease of most activity assays, it is logical to report carboxylesterase activity levels as well as other esterases (e.g., acetylcholinesterase). Although it is still unclear as to whether acetylcholinesterase or carboxylesterase is the most "appropriate" biomarker, there are sufficient data to suggest that at the very least further studies should be performed with carboxylesterases. Most likely, data will show that it is optimal to measure activity for both enzymes whenever possible. Acetylcholinesterase has the distinct advantage of a clear biological function, whereas the endogenous role of carboxylesterases is still unclear. However, a combination of activity measurements for the two enzyme systems will provide a much more detailed picture of organism health and insecticide exposure. The main outstanding issues are the choice of substrate for activity assays and which tissues/organisms are most appropriate for monitoring studies. Substrate choice is very important, because carboxylesterase activity consists of multiple isozymes that most likely fluctuate on an organism- and tissue-specific basis. It is therefore difficult to compare work in one organism with a specific substrate with work performed in a different organism with a different substrate. An attempt should therefore be made to standardize the method. The most logical choice is PNPA (p-nitrophenyl acetate), as this substrate is commercially available, requires inexpensive optics for assay measurements, and has been used extensively in the literature. However, none of these beneficial properties indicates that the substrate is an appropriate surrogate for a specific compound, e.g., pyrethroid-hydrolyzing activity. It will most likely be necessary to have more specific surrogate substrates for use in assays that require information on the ability to detoxify/hydrolyze specific environmental contaminants. The use of carboxylesterase activity in TIE protocols appears to have excellent promise, but there are further technical issues that should be addressed to increase the utility of the method. The main concerns include the large amount of nonspecific protein added to the testing system, which can lead to undesirable side effects including nonspecific reductions in observed toxicity, decrease in dissolved oxygen content, and organism growth. It is probable that these issues can be resolved with further assay development. The ideal solution would be to have a commercial recombinant carboxylesterase that possessed elevated pyrethroid-hydrolysis activity and which was readily available, homogeneous, and inexpensive. The availability of such an enzyme would address nearly all the current method shortcomings. Such a preparation would be extremely useful for the aquatic toxicology community. Further work should focus on screening available esterases for stability, cost, and activity on pyrethroids, with specific focus on esterases capable of distinguishing type I from type II pyrethroids. It would also be beneficial to identify esterases that are not sensitive to OP insecticides. Many esterases and lipases are available as sets to test chemical reactions for green chemistry, enabling large-scale screening. Other potential approaches to increase the utility of the enzyme include derivatization with polyethylene glycol (PEG) or cyanuric acid chloride to increase stability and reduce microbial degradation. It is also possible that the enzyme could be formulated in a sol gel preparation to increase stability. It is likely that the use of carboxylesterase addition will increase for applications in sediment TIEs. Carboxylesterases are an interesting and useful enzyme family that deserves further study for applications in environmental monitoring as well as to increase our understanding of the fundamental biological role(s) of these enzymes. There are, of course, other enzymes that show high esterase activity on pyrethroids but are not technically carboxylesterases in the alpha/beta-hydrolase fold protein family. These enzymes should also be examined for use in TIE protocols and "esterase" arrays as well as for general applications in environmental monitoring. One can envision the creation of a standardized screen of enzymes with esterase activity to (1) identify environmental contaminants, (2) estimate the potential toxic effects of new compounds on a range of organisms, and (3) monitor organism exposure to agrochemicals (and potentially other contaminants). This approach would provide a multibiomarker integrative assessment of esterase-inhibiting potential of a compound or mixture. In conclusion, much is still unknown about this enzyme family, indicating that this area is still wide open to researchers interested in the applications of carboxylesterase activity as well as basic biological questions into the nature of enzyme activity and the endogenous role of the enzyme.
Collapse
Affiliation(s)
- Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles vãg 2, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|