1
|
Stigler L, Köhler A, Koller M, Job L, Escher B, Potschka H, Thiermann H, Skerra A, Worek F, Wille T. Post-VX exposure treatment of rats with engineered phosphotriesterases. Arch Toxicol 2021; 96:571-583. [PMID: 34962578 PMCID: PMC8837561 DOI: 10.1007/s00204-021-03199-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/09/2021] [Indexed: 12/05/2022]
Abstract
The biologically stable and highly toxic organophosphorus nerve agent (OP) VX poses a major health threat. Standard medical therapy, consisting of reactivators and competitive muscarinic receptor antagonists, is insufficient. Recently, two engineered mutants of the Brevundimonas diminuta phosphotriesterase (PTE) with enhanced catalytic efficiency (kcat/KM = 21 to 38 × 106 M−1 min−1) towards VX and a preferential hydrolysis of the more toxic P(−) enantiomer were described: PTE-C23(R152E)-PAS(100)-10-2-C3(I106A/C59V/C227V/E71K)-PAS(200) (PTE-2), a single-chain bispecific enzyme with a PAS linker and tag having enlarged substrate spectrum, and 10-2-C3(C59V/C227V)-PAS(200) (PTE-3), a stabilized homodimeric enzyme with a double PASylation tag (PAS-tag) to reduce plasma clearance. To assess in vivo efficacy, these engineered enzymes were tested in an anesthetized rat model post-VX exposure (~ 2LD50) in comparison with the recombinant wild-type PTE (PTE-1), dosed at 1.0 mg kg−1 i.v.: PTE-2 dosed at 1.3 mg kg−1 i.v. (PTE-2.1) and 2.6 mg kg−1 i.v. (PTE-2.2) and PTE-3 at 1.4 mg kg−1 i.v. Injection of the mutants PTE-2.2 and PTE-3, 5 min after s.c. VX exposure, ensured survival and prevented severe signs of a cholinergic crisis. Inhibition of erythrocyte acetylcholinesterase (AChE) could not be prevented. However, medulla oblongata and diaphragm AChE activity was partially preserved. All animals treated with the wild-type enzyme, PTE-1, showed severe cholinergic signs and died during the observation period of 180 min. PTE-2.1 resulted in the survival of all animals, yet accompanied by severe signs of OP poisoning. This study demonstrates for the first time efficient detoxification in vivo achieved with low doses of heterodimeric PTE-2 as well as PTE-3 and indicates the suitability of these engineered enzymes for the development of highly effective catalytic scavengers directed against VX.
Collapse
Affiliation(s)
- Lisa Stigler
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Anja Köhler
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany.,Chair of Biological Chemistry, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354, Freising, Germany
| | - Marianne Koller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Laura Job
- Chair of Biological Chemistry, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354, Freising, Germany
| | - Benjamin Escher
- Chair of Biological Chemistry, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354, Freising, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University Munich, Königinstraße 16, 80539, Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Arne Skerra
- Chair of Biological Chemistry, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354, Freising, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany.
| |
Collapse
|
2
|
Köhler A, Escher B, Job L, Koller M, Thiermann H, Skerra A, Worek F. Catalytic activity and stereoselectivity of engineered phosphotriesterases towards structurally different nerve agents in vitro. Arch Toxicol 2021; 95:2815-2823. [PMID: 34160649 PMCID: PMC8298220 DOI: 10.1007/s00204-021-03094-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Highly toxic organophosphorus nerve agents, especially the extremely stable and persistent V-type agents such as VX, still pose a threat to the human population and require effective medical countermeasures. Engineered mutants of the Brevundimonas diminuta phosphotriesterase (BdPTE) exhibit enhanced catalytic activities and have demonstrated detoxification in animal models, however, substrate specificity and fast plasma clearance limit their medical applicability. To allow better assessment of their substrate profiles, we have thoroughly investigated the catalytic efficacies of five BdPTE mutants with 17 different nerve agents using an AChE inhibition assay. In addition, we studied one BdPTE version that was fused with structurally disordered PAS polypeptides to enable delayed plasma clearance and one bispecific BdPTE with broadened substrate spectrum composed of two functionally distinct subunits connected by a PAS linker. Measured kcat/KM values were as high as 6.5 and 1.5 × 108 M-1 min-1 with G- and V-agents, respectively. Furthermore, the stereoselective degradation of VX enantiomers by the PASylated BdPTE-4 and the bispecific BdPTE-7 were investigated by chiral LC-MS/MS, resulting in a several fold faster hydrolysis of the more toxic P(-) VX stereoisomer compared to P(+) VX. In conclusion, the newly developed enzymes BdPTE-4 and BdPTE-7 have shown high catalytic efficacy towards structurally different nerve agents and stereoselectivity towards the toxic P(-) VX enantiomer in vitro and offer promise for use as bioscavengers in vivo.
Collapse
Affiliation(s)
- Anja Köhler
- Institut für Pharmakologie und Toxikologie der Bundeswehr, 80937, Munich, Germany
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising, Germany
| | - Benjamin Escher
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising, Germany
| | - Laura Job
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising, Germany
| | - Marianne Koller
- Institut für Pharmakologie und Toxikologie der Bundeswehr, 80937, Munich, Germany
| | - Horst Thiermann
- Institut für Pharmakologie und Toxikologie der Bundeswehr, 80937, Munich, Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354, Freising, Germany.
| | - Franz Worek
- Institut für Pharmakologie und Toxikologie der Bundeswehr, 80937, Munich, Germany.
| |
Collapse
|
3
|
Hrvat NM, Kovarik Z. Counteracting poisoning with chemical warfare nerve agents. Arh Hig Rada Toksikol 2020; 71:266-284. [PMID: 33410774 PMCID: PMC7968514 DOI: 10.2478/aiht-2020-71-3459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/01/2020] [Accepted: 11/01/2020] [Indexed: 12/14/2022] Open
Abstract
Phosphylation of the pivotal enzyme acetylcholinesterase (AChE) by nerve agents (NAs) leads to irreversible inhibition of the enzyme and accumulation of neurotransmitter acetylcholine, which induces cholinergic crisis, that is, overstimulation of muscarinic and nicotinic membrane receptors in the central and peripheral nervous system. In severe cases, subsequent desensitisation of the receptors results in hypoxia, vasodepression, and respiratory arrest, followed by death. Prompt action is therefore critical to improve the chances of victim's survival and recovery. Standard therapy of NA poisoning generally involves administration of anticholinergic atropine and an oxime reactivator of phosphylated AChE. Anticholinesterase compounds or NA bioscavengers can also be applied to preserve native AChE from inhibition. With this review of 70 years of research we aim to present current and potential approaches to counteracting NA poisoning.
Collapse
Affiliation(s)
| | - Zrinka Kovarik
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| |
Collapse
|
4
|
Escher B, Köhler A, Job L, Worek F, Skerra A. Translating the Concept of Bispecific Antibodies to Engineering Heterodimeric Phosphotriesterases with Broad Organophosphate Substrate Recognition. Biochemistry 2020; 59:4395-4406. [PMID: 33146522 DOI: 10.1021/acs.biochem.0c00751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We have adopted the concept of bispecific antibodies, which can simultaneously block or cross-link two different biomolecular targets, to create bispecific enzymes by exploiting the homodimeric quaternary structure of bacterial phosphotriesterases (PTEs). The PTEs from Brevundimonas diminuta and Agrobacterium radiobacter, whose engineered variants can efficiently hydrolyze organophosphorus (OP) nerve agents and pesticides, respectively, have attracted considerable interest for the treatment of the corresponding intoxications. OP nerve agents and pesticides still pose a severe toxicological threat in military conflicts, including acts of terrorism, as well as in agriculture, leading to >100000 deaths per year. In principle, engineered conventional homodimeric PTEs may provoke hydrolytic inactivation of individual OPs in vivo, and their application as catalytic bioscavengers via administration into the bloodstream has been proposed. However, their narrow substrate specificity would necessitate therapeutic application of a set or mixture of different enzymes, which complicates biopharmaceutical development. We succeeded in combining subunits from both enzymes and to stabilize their heterodimerization by rationally designing electrostatic steering mutations, thus breaking the natural C2 symmetry. The resulting bispecific enzyme from two PTEs with different bacterial origin exhibits an ultrabroad OP substrate profile and allows the efficient detoxification of both nerve agents and pesticides. Our approach of combining two active sites with distinct substrate specificities within one artificial dimeric biocatalyst-retaining the size and general properties of the original enzyme without utilizing protein mixtures or much larger fusion proteins-not only should facilitate biological drug development but also may be applicable to oligomeric enzymes with other catalytic activities.
Collapse
Affiliation(s)
- Benjamin Escher
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Anja Köhler
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany.,Bundeswehr Institut für Pharmakologie und Toxikologie, Neuherbergstrasse 11, 80937 München, Germany
| | - Laura Job
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Franz Worek
- Bundeswehr Institut für Pharmakologie und Toxikologie, Neuherbergstrasse 11, 80937 München, Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| |
Collapse
|
5
|
Despotović D, Aharon E, Dubovetskyi A, Leader H, Ashani Y, Tawfik DS. A mixture of three engineered phosphotriesterases enables rapid detoxification of the entire spectrum of known threat nerve agents. Protein Eng Des Sel 2020; 32:169-174. [PMID: 31612205 DOI: 10.1093/protein/gzz039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 01/22/2023] Open
Abstract
Nerve agents are organophosphates (OPs) that potently inhibit acetylcholinesterase, and their enzymatic detoxification has been a long-standing goal. Nerve agents vary widely in size, charge, hydrophobicity and the cleavable ester bond. A single enzyme is therefore unlikely to efficiently hydrolyze all agents. Here, we describe a mixture of three previously developed variants of the bacterial phosphotriesterase (Bd-PTE) that are highly stable and nearly sequence identical. This mixture enables effective detoxification of a broad spectrum of known threat agents-GA (tabun), GB (sarin), GD (soman), GF (cyclosarin), VX and Russian-VX. The potential for dimer dissociation and exchange that could inactivate Bd-PTE has minimal impact, and the three enzyme variants are as active in a mixture as they are individually. To our knowledge, this engineered enzyme 'cocktail' comprises the first solution for enzymatic detoxification of the entire range of threat nerve agents.
Collapse
Affiliation(s)
- Dragana Despotović
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| | - Einav Aharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| | - Artem Dubovetskyi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| | - Haim Leader
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| | - Yacov Ashani
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| | - Dan S Tawfik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Herzl st. 234, Rehovot 7610001, Israel
| |
Collapse
|
6
|
Targeting organophosphorus compounds poisoning by novel quinuclidine-3 oximes: development of butyrylcholinesterase-based bioscavengers. Arch Toxicol 2020; 94:3157-3171. [PMID: 32583098 DOI: 10.1007/s00204-020-02811-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/15/2020] [Indexed: 01/04/2023]
Abstract
A library of 14 mono-oxime quinuclidinium-based compounds with alkyl or benzyl substituent were synthesized and characterized in vitro as potential antidotes for organophosphorus compounds (OP) poisoning treatment. We evaluated their potency for reversible inhibition and reactivation of OP inhibited human acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) and evaluated interactions by molecular docking studies. The reactivation was notable for both AChE and BChE inhibited by VX, cyclosarin, sarin and paraoxon, if quinuclidinium compounds contained the benzyl group attached to the quinuclidinium moiety. Out of all 14, oxime Q8 [4-bromobenzyl-3-(hydroxyimino)quinuclidinium bromide] was singled out as having the highest determined overall reactivation rate of approximately 20,000 M-1 min-1 for cyclosarin-inhibited BChE. Furthermore, this oxime in combination with BChE exhibited a capability to act as a bioscavenger of cyclosarin, degrading within 2 h up to 100-fold excess of cyclosarin concentration over the enzyme. Molecular modeling revealed that the position of the cyclohexyl moiety conjugated with the active site serine of BChE directs the favorable positioning of the quinuclidinium ring and the bromophenyl moiety of Q8, which makes phosphonylated-serine easily accessible for the nucleophilic displacement by the oxime group of Q8. This result presents a novel scaffold for the development of new BChE-based bioscavengers. Furthermore, a cytotoxic effect was not observed for Q8, which also makes it promising for further in vivo reactivation studies.
Collapse
|
7
|
Betapudi V, Goswami R, Silayeva L, Doctor DM, Chilukuri N. Gene therapy delivering a paraoxonase 1 variant offers long-term prophylactic protection against nerve agents in mice. Sci Transl Med 2020; 12:12/527/eaay0356. [PMID: 31969483 DOI: 10.1126/scitranslmed.aay0356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/19/2019] [Accepted: 11/13/2019] [Indexed: 11/02/2022]
Abstract
Chemical warfare nerve agents are organophosphorus chemical compounds that induce cholinergic crisis, leaving little or no time for medical intervention to prevent death. The current chemical treatment regimen may prevent death but does not prevent postexposure complications such as brain damage and permanent behavioral abnormalities. In the present study, we have demonstrated an adeno-associated virus 8 (AAV8)-mediated paraoxonase 1 variant IF-11 (PON1-IF11) gene therapy that offers asymptomatic prophylactic protection to mice against multiple lethal doses of G-type chemical warfare nerve agents, namely, tabun, sarin, cyclosarin, and soman, for up to 5 months in mice. A single injection of liver-specific adeno-associated viral particles loaded with PON1-IF11 gene resulted in expression and secretion of recombinant PON1-IF11 in milligram quantities, which has the catalytic power to break down G-type chemical warfare nerve agents into biologically inactive products in vitro and in vivo in rodents. Mice containing milligram concentrations of recombinant PON1-IF11 in their blood displayed no clinical signs of toxicity, as judged by their hematological parameters and serum chemistry profiles. Our study unfolds avenues to develop a one-time application of gene therapy to express a near-natural and circulating therapeutic PON1-IF11 protein that can potentially protect humans against G-type chemical warfare nerve agents for several weeks to months.
Collapse
Affiliation(s)
- Venkaiah Betapudi
- Medical Toxicology Research Division, Biochemistry & Physiology Department, Agent Mitigation, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Reena Goswami
- Medical Toxicology Research Division, Biochemistry & Physiology Department, Agent Mitigation, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Liliya Silayeva
- Medical Toxicology Research Division, Biochemistry & Physiology Department, Agent Mitigation, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Deborah M Doctor
- Medical Toxicology Research Division, Biochemistry & Physiology Department, Agent Mitigation, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Nageswararao Chilukuri
- Medical Toxicology Research Division, Biochemistry & Physiology Department, Agent Mitigation, United States Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010-5400, USA.
| |
Collapse
|
8
|
Catalytic bioscavengers as countermeasures against organophosphate nerve agents. Chem Biol Interact 2018; 292:50-64. [DOI: 10.1016/j.cbi.2018.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
|
9
|
Lushchekina SV, Schopfer LM, Grigorenko BL, Nemukhin AV, Varfolomeev SD, Lockridge O, Masson P. Optimization of Cholinesterase-Based Catalytic Bioscavengers Against Organophosphorus Agents. Front Pharmacol 2018; 9:211. [PMID: 29593539 PMCID: PMC5859046 DOI: 10.3389/fphar.2018.00211] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
Organophosphorus agents (OPs) are irreversible inhibitors of acetylcholinesterase (AChE). OP poisoning causes major cholinergic syndrome. Current medical counter-measures mitigate the acute effects but have limited action against OP-induced brain damage. Bioscavengers are appealing alternative therapeutic approach because they neutralize OPs in bloodstream before they reach physiological targets. First generation bioscavengers are stoichiometric bioscavengers. However, stoichiometric neutralization requires administration of huge doses of enzyme. Second generation bioscavengers are catalytic bioscavengers capable of detoxifying OPs with a turnover. High bimolecular rate constants (kcat/Km > 106 M−1min−1) are required, so that low enzyme doses can be administered. Cholinesterases (ChE) are attractive candidates because OPs are hemi-substrates. Moderate OP hydrolase (OPase) activity has been observed for certain natural ChEs and for G117H-based human BChE mutants made by site-directed mutagenesis. However, before mutated ChEs can become operational catalytic bioscavengers their dephosphylation rate constant must be increased by several orders of magnitude. New strategies for converting ChEs into fast OPase are based either on combinational approaches or on computer redesign of enzyme. The keystone for rational conversion of ChEs into OPases is to understand the reaction mechanisms with OPs. In the present work we propose that efficient OP hydrolysis can be achieved by re-designing the configuration of enzyme active center residues and by creating specific routes for attack of water molecules and proton transfer. Four directions for nucleophilic attack of water on phosphorus atom were defined. Changes must lead to a novel enzyme, wherein OP hydrolysis wins over competing aging reactions. Kinetic, crystallographic, and computational data have been accumulated that describe mechanisms of reactions involving ChEs. From these studies, it appears that introducing new groups that create a stable H-bonded network susceptible to activate and orient water molecule, stabilize transition states (TS), and intermediates may determine whether dephosphylation is favored over aging. Mutations on key residues (L286, F329, F398) were considered. QM/MM calculations suggest that mutation L286H combined to other mutations favors water attack from apical position. However, the aging reaction is competing. Axial direction of water attack is not favorable to aging. QM/MM calculation shows that F329H+F398H-based multiple mutants display favorable energy barrier for fast reactivation without aging.
Collapse
Affiliation(s)
- Sofya V Lushchekina
- Laboratory of Computer Modeling of Bimolecular Systems and Nanomaterials, N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia
| | - Lawrence M Schopfer
- Department of Biochemistry and Molecular Biology, Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Bella L Grigorenko
- Laboratory of Computer Modeling of Bimolecular Systems and Nanomaterials, N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia.,Chemistry Department, Lomonosov State University, Moscow, Russia
| | - Alexander V Nemukhin
- Laboratory of Computer Modeling of Bimolecular Systems and Nanomaterials, N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia.,Chemistry Department, Lomonosov State University, Moscow, Russia
| | - Sergei D Varfolomeev
- Laboratory of Computer Modeling of Bimolecular Systems and Nanomaterials, N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, Russia.,Chemistry Department, Lomonosov State University, Moscow, Russia
| | - Oksana Lockridge
- Department of Biochemistry and Molecular Biology, Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Patrick Masson
- Neuropharmacology Laboratory, Kazan Federal University, Kazan, Russia
| |
Collapse
|
10
|
Masson P, Nachon F. Cholinesterase reactivators and bioscavengers for pre- and post-exposure treatments of organophosphorus poisoning. J Neurochem 2017; 142 Suppl 2:26-40. [PMID: 28542985 DOI: 10.1111/jnc.14026] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/02/2017] [Accepted: 03/10/2017] [Indexed: 12/11/2022]
Abstract
Organophosphorus agents (OPs) irreversibly inhibit acetylcholinesterase (AChE) causing a major cholinergic syndrome. The medical counter-measures of OP poisoning have not evolved for the last 30 years with carbamates for pretreatment, pyridinium oximes-based AChE reactivators, antimuscarinic drugs and neuroprotective benzodiazepines for post-exposure treatment. These drugs ensure protection of peripheral nervous system and mitigate acute effects of OP lethal doses. However, they have significant limitations. Pyridostigmine and oximes do not protect/reactivate central AChE. Oximes poorly reactivate AChE inhibited by phosphoramidates. In addition, current neuroprotectants do not protect the central nervous system shortly after the onset of seizures when brain damage becomes irreversible. New therapeutic approaches for pre- and post-exposure treatments involve detoxification of OP molecules before they reach their molecular targets by administrating catalytic bioscavengers, among them phosphotriesterases are the most promising. Novel generation of broad spectrum reactivators are designed for crossing the blood-brain barrier and reactivate central AChE. This is an article for the special issue XVth International Symposium on Cholinergic Mechanisms.
Collapse
Affiliation(s)
- Patrick Masson
- Neuropharmacology Laboratory, Kazan Federal University, Kazan, Russia
| | - Florian Nachon
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, Cédex, France
| |
Collapse
|
11
|
Langston JL, Myers TM. VX toxicity in the Göttingen minipig. Toxicol Lett 2016; 264:12-19. [PMID: 27773723 DOI: 10.1016/j.toxlet.2016.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/20/2016] [Accepted: 10/19/2016] [Indexed: 11/18/2022]
Abstract
The present experiments determined the intramuscular LD50 of VX in male Göttingen minipigs at two stages of development. In pubertal animals (115 days old), the LD50 of VX was indeterminate, but approximated 33.3μg/kg. However, in sexually mature animals (152 days old), the LD50 was estimated to be only 17.4μg/kg. Signs of nerve agent toxicity in the Göttingen minipig were similar to those described for other species, with some notable exceptions (such as urticaria and ejaculation). Latencies to the onset of sustained convulsions were inversely related to the administered dose of VX in both ages of minipigs. Additionally, actigraphy was used to quantify the presence of tremor and convulsions and, in some cases, was useful for precisely estimating time of death. The main finding indicates that in minipigs, as in other species, even relatively small differences in age can substantially alter the toxicity of nerve agents. Additionally, actigraphy can serve as a non-invasive method of characterizing the tremors and convulsions that often accompany nerve agent intoxication.
Collapse
Affiliation(s)
- Jeffrey L Langston
- Analytical Toxicology Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Todd M Myers
- Analytical Toxicology Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA.
| |
Collapse
|