Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(9): 6333-6353, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37094110

ABSTRACT

Insecticide resistance jeopardizes the prevention of infectious diseases such as malaria and dengue fever by vector control of disease-transmitting mosquitoes. Effective new insecticidal compounds with minimal adverse effects on humans and the environment are therefore urgently needed. Here, we explore noncovalent inhibitors of the well-validated insecticidal target acetylcholinesterase (AChE) based on a 4-thiazolidinone scaffold. The 4-thiazolidinones inhibit AChE1 from the mosquitoes Anopheles gambiae and Aedes aegypti at low micromolar concentrations. Their selectivity depends primarily on the substitution pattern of the phenyl ring; halogen substituents have complex effects. The compounds also feature a pendant aliphatic amine that was important for activity; little variation of this group is tolerated. Molecular docking studies suggested that the tight selectivity profiles of these compounds are due to competition between two binding sites. Three 4-thiazolidinones tested for in vivo insecticidal activity had similar effects on disease-transmitting mosquitoes despite a 10-fold difference in their in vitro activity.


Subject(s)
Aedes , Anopheles , Insecticides , Animals , Humans , Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Molecular Docking Simulation , Mosquito Vectors , Insecticides/pharmacology , Insecticides/chemistry , Structure-Activity Relationship
2.
Chemistry ; 28(40): e202200678, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35420233

ABSTRACT

Reactivators are vital for the treatment of organophosphorus nerve agent (OPNA) intoxication but new alternatives are needed due to their limited clinical applicability. The toxicity of OPNAs stems from covalent inhibition of the essential enzyme acetylcholinesterase (AChE), which reactivators relieve via a chemical reaction with the inactivated enzyme. Here, we present new strategies and tools for developing reactivators. We discover suitable inhibitor scaffolds by using an activity-independent competition assay to study non-covalent interactions with OPNA-AChEs and transform these inhibitors into broad-spectrum reactivators. Moreover, we identify determinants of reactivation efficiency by analysing reactivation and pre-reactivation kinetics together with structural data. Our results show that new OPNA reactivators can be discovered rationally by exploiting detailed knowledge of the reactivation mechanism of OPNA-inhibited AChE.


Subject(s)
Cholinesterase Reactivators , Nerve Agents , Acetylcholinesterase/chemistry , Antidotes , Cholinesterase Inhibitors/pharmacology , Cholinesterase Reactivators/chemistry , Organophosphorus Compounds , Oximes/chemistry
3.
ACS Med Chem Lett ; 13(3): 499-506, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35300078

ABSTRACT

Multitarget directed ligands (MTDLs) represent a promising frontier in tackling the complexity of multifactorial pathologies. The synergistic inhibition of monoamine oxidase B (MAO B) and acetylcholinesterase (AChE) is believed to provide a potentiated effect in the treatment of Alzheimer's disease. Among previously reported micromolar or sub-micromolar coumarin-bearing dual inhibitors, compound 1 returned a tight-binding inhibition of MAO B (K i = 4.5 µM) and a +5.5 °C increase in the enzyme T m value. Indeed, the X-ray crystal structure revealed that binding of 1 produces unforeseen conformational changes at the MAO B entrance cavity. Interestingly, 1 showed great shape complementarity with the AChE enzymatic gorge, being deeply buried from the catalytic anionic subsite (CAS) to the peripheral anionic subsite (PAS) and causing significant structural changes in the active site. These findings provide structural templates for further development of dual MAO B and AChE inhibitors.

4.
Angew Chem Int Ed Engl ; 60(2): 813-819, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33079431

ABSTRACT

The potential drug target choline acetyltransferase (ChAT) catalyses the production of the neurotransmitter acetylcholine in cholinergic neurons, T-cells, and B-cells. Herein, we show that arylvinylpyridiniums (AVPs), the most widely studied class of ChAT inhibitors, act as substrate in an unusual coenzyme A-dependent hydrothiolation reaction. This in situ synthesis yields an adduct that is the actual enzyme inhibitor. The adduct is deeply buried in the active site tunnel of ChAT and interactions with a hydrophobic pocket near the choline binding site have major implications for the molecular recognition of inhibitors. Our findings clarify the inhibition mechanism of AVPs, establish a drug modality that exploits a target-catalysed reaction between exogenous and endogenous precursors, and provide new directions for the development of ChAT inhibitors with improved potency and bioactivity.


Subject(s)
Choline O-Acetyltransferase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Ligands , Acetylcholine/metabolism , Binding Sites , Biocatalysis , Catalytic Domain , Choline O-Acetyltransferase/metabolism , Enzyme Inhibitors/metabolism , Kinetics , Molecular Dynamics Simulation , Pyridines/chemistry , Pyridines/metabolism , Thermodynamics , Transition Temperature
5.
J Phys Chem B ; 124(30): 6529-6539, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32610016

ABSTRACT

Arene-arene interactions play important roles in protein-ligand complex formation. Here, we investigate the characteristics of arene-arene interactions between small organic molecules and aromatic amino acids in protein interiors. The study is based on X-ray crystallographic data and quantum mechanical calculations using the enzyme acetylcholinesterase and selected inhibitory ligands as a model system. It is shown that the arene substituents of the inhibitors dictate the strength of the interaction and the geometry of the resulting complexes. Importantly, the calculated interaction energies correlate well with the measured inhibitor potency. Non-hydrogen substituents strengthened all interaction types in the protein milieu, in keeping with results for benzene dimer model systems. The interaction energies were dispersion-dominated, but substituents that induced local dipole moments increased the electrostatic contribution and thus yielded more strongly bound complexes. These findings provide fundamental insights into the physical mechanisms governing arene-arene interactions in the protein milieu and thus into molecular recognition between proteins and small molecules.


Subject(s)
Benzene , Crystallography, X-Ray , Ligands , Static Electricity
6.
J Med Chem ; 61(23): 10545-10557, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30339371

ABSTRACT

Resistance development in insects significantly threatens the important benefits obtained by insecticide usage in vector control of disease-transmitting insects. Discovery of new chemical entities with insecticidal activity is highly desired in order to develop new insecticide candidates. Here, we present the design, synthesis, and biological evaluation of phenoxyacetamide-based inhibitors of the essential enzyme acetylcholinesterase 1 (AChE1). AChE1 is a validated insecticide target to control mosquito vectors of, e.g., malaria, dengue, and Zika virus infections. The inhibitors combine a mosquito versus human AChE selectivity with a high potency also for the resistance-conferring mutation G122S; two properties that have proven challenging to combine in a single compound. Structure-activity relationship analyses and molecular dynamics simulations of inhibitor-protein complexes have provided insights that elucidate the molecular basis for these properties. We also show that the inhibitors demonstrate in vivo insecticidal activity on disease-transmitting mosquitoes. Our findings support the concept of noncovalent, selective, and resistance-breaking inhibitors of AChE1 as a promising approach for future insecticide development.


Subject(s)
Acetylcholinesterase/metabolism , Aedes/enzymology , Cholinesterase Inhibitors/pharmacology , Drug Resistance/drug effects , Acetamides/chemistry , Acetamides/pharmacology , Acetylcholinesterase/chemistry , Animals , Cholinesterase Inhibitors/chemistry , Drug Design , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Protein Conformation
7.
J Phys Chem B ; 122(36): 8516-8525, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30110543

ABSTRACT

The enzyme acetylcholinesterase (AChE) is essential in humans and animals because it catalyzes the breakdown of the nerve-signaling substance acetylcholine. Small molecules that inhibit the function of AChE are important for their use as drugs in the, for example, symptomatic treatment of Alzheimer's disease. New and improved inhibitors are warranted, mainly because of severe side effects of current drugs. In the present study, we have investigated if and how two enantiomeric inhibitors of AChE influence the overall dynamics of noncovalent complexes, using elastic incoherent neutron scattering. A fruitful combination of univariate models, including a newly developed non-Gaussian model for atomic fluctuations, and multivariate methods (principal component analysis and discriminant analysis) was crucial to analyze the fine details of the data. The study revealed a small but clear increase in the dynamics of the inhibited enzyme compared to that of the noninhibited enzyme and contributed to the fundamental knowledge of the mechanisms of AChE-inhibitor binding valuable for the future development of inhibitors.


Subject(s)
Acetylcholinesterase/chemistry , Benzamides/chemistry , Cholinesterase Inhibitors/chemistry , Acetylcholinesterase/metabolism , Animals , Benzamides/metabolism , Cholinesterase Inhibitors/metabolism , Mice , Multivariate Analysis , Neutron Diffraction , Protein Binding , Protein Conformation , Stereoisomerism
8.
Molecules ; 22(9)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28867801

ABSTRACT

Acetylcholinesterase (AChE) is an essential enzyme that terminates cholinergic transmission by a rapid hydrolysis of the neurotransmitter acetylcholine. AChE is an important target for treatment of various cholinergic deficiencies, including Alzheimer's disease and myasthenia gravis. In a previous high throughput screening campaign, we identified the dye crystal violet (CV) as an inhibitor of AChE. Herein, we show that CV displays a significant cooperativity for binding to AChE, and the molecular basis for this observation has been investigated by X-ray crystallography. Two monomers of CV bind to residues at the entrance of the active site gorge of the enzyme. Notably, the two CV molecules have extensive intermolecular contacts with each other and with AChE. Computational analyses show that the observed CV dimer is not stable in solution, suggesting the sequential binding of two monomers. Guided by the structural analysis, we designed a set of single site substitutions, and investigated their effect on the binding of CV. Only moderate effects on the binding and the cooperativity were observed, suggesting a robustness in the interaction between CV and AChE. Taken together, we propose that the dimeric cooperative binding is due to a rare combination of chemical and structural properties of both CV and the AChE molecule itself.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Dimerization , Gentian Violet/pharmacology , Acetylcholinesterase/chemistry , Animals , Binding Sites , Cholinesterase Inhibitors/chemistry , Computer Simulation , Crystallography, X-Ray , Gentian Violet/chemistry , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Static Electricity
9.
Eur J Med Chem ; 134: 415-427, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28433681

ABSTRACT

Vector control of disease-transmitting mosquitoes by insecticides has a central role in reducing the number of parasitic- and viral infection cases. The currently used insecticides are efficient, but safety concerns and the development of insecticide-resistant mosquito strains warrant the search for alternative compound classes for vector control. Here, we have designed and synthesized thiourea-based compounds as non-covalent inhibitors of acetylcholinesterase 1 (AChE1) from the mosquitoes Anopheles gambiae (An. gambiae) and Aedes aegypti (Ae. aegypti), as well as a naturally occurring resistant-conferring mutant. The N-aryl-N'-ethyleneaminothioureas proved to be inhibitors of AChE1; the most efficient one showed submicromolar potency. Importantly, the inhibitors exhibited selectivity over the human AChE (hAChE), which is desirable for new insecticides. The structure-activity relationship (SAR) analysis of the thioureas revealed that small changes in the chemical structure had a large effect on inhibition capacity. The thioureas showed to have different SAR when inhibiting AChE1 and hAChE, respectively, enabling an investigation of structure-selectivity relationships. Furthermore, insecticidal activity was demonstrated using adult and larvae An. gambiae and Ae. aegypti mosquitoes.


Subject(s)
Aedes/drug effects , Anopheles/drug effects , Cholinesterase Inhibitors/toxicity , Insect Vectors/drug effects , Insecticides/toxicity , Thiourea/toxicity , Acetylcholinesterase/metabolism , Aedes/enzymology , Animals , Anopheles/enzymology , Cholinesterase Inhibitors/chemistry , Female , Humans , Insect Proteins/antagonists & inhibitors , Insect Proteins/metabolism , Insect Vectors/enzymology , Insecticides/chemistry , Larva/drug effects , Larva/enzymology , Thiourea/analogs & derivatives
10.
J Med Chem ; 59(20): 9409-9421, 2016 Oct 27.
Article in English | MEDLINE | ID: mdl-27598521

ABSTRACT

Vector control of disease-transmitting mosquitoes is increasingly important due to the re-emergence and spread of infections such as malaria and dengue. We have conducted a high throughput screen (HTS) of 17,500 compounds for inhibition of the essential AChE1 enzymes from the mosquitoes Anopheles gambiae and Aedes aegypti. In a differential HTS analysis including the human AChE, several structurally diverse, potent, and selective noncovalent AChE1 inhibitors were discovered. For example, a phenoxyacetamide-based inhibitor was identified with a 100-fold selectivity for the mosquito over the human enzyme. The compound also inhibited a resistance conferring mutant of AChE1. Structure-selectivity relationships could be proposed based on the enzymes' 3D structures; the hits' selectivity profiles appear to be linked to differences in two loops that affect the structure of the entire active site. Noncovalent inhibitors of AChE1, such as the ones presented here, provide valuable starting points toward insecticides and are complementary to existing and new covalent inhibitors.


Subject(s)
Acetylcholinesterase/metabolism , Aedes/drug effects , Anopheles/drug effects , Cholinesterase Inhibitors/pharmacology , Drug Discovery , Insect Vectors/drug effects , Insect Vectors/enzymology , Insecticides/pharmacology , Acetylcholinesterase/genetics , Aedes/enzymology , Animals , Anopheles/enzymology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Insecticides/chemical synthesis , Insecticides/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
11.
Proc Natl Acad Sci U S A ; 113(20): 5514-9, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140636

ABSTRACT

Organophosphorus nerve agents interfere with cholinergic signaling by covalently binding to the active site of the enzyme acetylcholinesterase (AChE). This inhibition causes an accumulation of the neurotransmitter acetylcholine, potentially leading to overstimulation of the nervous system and death. Current treatments include the use of antidotes that promote the release of functional AChE by an unknown reactivation mechanism. We have used diffusion trap cryocrystallography and density functional theory (DFT) calculations to determine and analyze prereaction conformers of the nerve agent antidote HI-6 in complex with Mus musculus AChE covalently inhibited by the nerve agent sarin. These analyses reveal previously unknown conformations of the system and suggest that the cleavage of the covalent enzyme-sarin bond is preceded by a conformational change in the sarin adduct itself. Together with data from the reactivation kinetics, this alternate conformation suggests a key interaction between Glu202 and the O-isopropyl moiety of sarin. Moreover, solvent kinetic isotope effect experiments using deuterium oxide reveal that the reactivation mechanism features an isotope-sensitive step. These findings provide insights into the reactivation mechanism and provide a starting point for the development of improved antidotes. The work also illustrates how DFT calculations can guide the interpretation, analysis, and validation of crystallographic data for challenging reactive systems with complex conformational dynamics.


Subject(s)
Acetylcholinesterase/chemistry , Antidotes/chemistry , Cholinesterase Reactivators/chemistry , Nerve Agents/chemistry , Oximes/chemistry , Pyridinium Compounds/chemistry , Sarin/chemistry , Crystallography, X-Ray , Kinetics , Molecular Conformation
12.
Chemistry ; 22(8): 2672-81, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26751405

ABSTRACT

Molecular recognition events in biological systems are driven by non-covalent interactions between interacting species. Here, we have studied hydrogen bonds of the CH⋅⋅⋅Y type involving electron-deficient CH donors using dispersion-corrected density functional theory (DFT) calculations applied to acetylcholinesterase-ligand complexes. The strengths of CH⋅⋅⋅Y interactions activated by a proximal cation were considerably strong; comparable to or greater than those of classical hydrogen bonds. Significant differences in the energetic components compared to classical hydrogen bonds and non-activated CH⋅⋅⋅Y interactions were observed. Comparison between DFT and molecular mechanics calculations showed that common force fields could not reproduce the interaction energy values of the studied hydrogen bonds. The presented results highlight the importance of considering CH⋅⋅⋅Y interactions when analysing protein-ligand complexes, call for a review of current force fields, and opens up possibilities for the development of improved design tools for drug discovery.


Subject(s)
Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Drug Discovery/methods , Hydrogen Bonding , Ligands , Models, Molecular , Quantum Theory
13.
PLoS One ; 10(10): e0138598, 2015.
Article in English | MEDLINE | ID: mdl-26447952

ABSTRACT

Mosquitoes of the Anopheles (An.) and Aedes (Ae.) genus are principal vectors of human diseases including malaria, dengue and yellow fever. Insecticide-based vector control is an established and important way of preventing transmission of such infections. Currently used insecticides can efficiently control mosquito populations, but there are growing concerns about emerging resistance, off-target toxicity and their ability to alter ecosystems. A potential target for the development of insecticides with reduced off-target toxicity is the cholinergic enzyme acetylcholinesterase (AChE). Herein, we report cloning, baculoviral expression and functional characterization of the wild-type AChE genes (ace-1) from An. gambiae and Ae. aegypti, including a naturally occurring insecticide-resistant (G119S) mutant of An. gambiae. Using enzymatic digestion and liquid chromatography-tandem mass spectrometry we found that the secreted proteins were post-translationally modified. The Michaelis-Menten constants and turnover numbers of the mosquito enzymes were lower than those of the orthologous AChEs from Mus musculus and Homo sapiens. We also found that the G119S substitution reduced the turnover rate of substrates and the potency of selected covalent inhibitors. Furthermore, non-covalent inhibitors were less sensitive to the G119S substitution and differentiate the mosquito enzymes from corresponding vertebrate enzymes. Our findings indicate that it may be possible to develop selective non-covalent inhibitors that effectively target both the wild-type and insecticide resistant mutants of mosquito AChE.


Subject(s)
Acetylcholinesterase/metabolism , Aedes/enzymology , Anopheles/enzymology , Ecosystem , Insect Vectors/enzymology , Acetylcholinesterase/genetics , Aedes/genetics , Animals , Anopheles/genetics , Anopheles/metabolism , Chromatography, Liquid , Insect Vectors/genetics , Malaria/prevention & control , Tandem Mass Spectrometry
14.
J Comput Aided Mol Des ; 29(3): 199-215, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25351962

ABSTRACT

Scientific disciplines such as medicinal- and environmental chemistry, pharmacology, and toxicology deal with the questions related to the effects small organic compounds exhort on biological targets and the compounds' physicochemical properties responsible for these effects. A common strategy in this endeavor is to establish structure-activity relationships (SARs). The aim of this work was to illustrate benefits of performing a statistical molecular design (SMD) and proper statistical analysis of the molecules' properties before SAR and quantitative structure-activity relationship (QSAR) analysis. Our SMD followed by synthesis yielded a set of inhibitors of the enzyme acetylcholinesterase (AChE) that had very few inherent dependencies between the substructures in the molecules. If such dependencies exist, they cause severe errors in SAR interpretation and predictions by QSAR-models, and leave a set of molecules less suitable for future decision-making. In our study, SAR- and QSAR models could show which molecular sub-structures and physicochemical features that were advantageous for the AChE inhibition. Finally, the QSAR model was used for the prediction of the inhibition of AChE by an external prediction set of molecules. The accuracy of these predictions was asserted by statistical significance tests and by comparisons to simple but relevant reference models.


Subject(s)
Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Models, Statistical , Quantitative Structure-Activity Relationship , Acetylcholinesterase/metabolism , Analysis of Variance , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/chemical synthesis , Models, Molecular , Molecular Structure , Structure-Activity Relationship
15.
Pestic Biochem Physiol ; 106(3): 79-84, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24003261

ABSTRACT

Conventional insecticides targeting acetylcholinesterase (AChE) typically show high mammalian toxicities and because there is resistance to these compounds in many insect species, alternatives to established AChE inhibitors used for pest control are needed. Here we used a fluorescence method to monitor interactions between various AChE inhibitors and the AChE peripheral anionic site, which is a novel target for new insecticides acting on this enzyme. The assay uses thioflavin-T as a probe, which binds to the peripheral anionic site of AChE and yields an increase in fluorescent signal. Three types of AChE inhibitors were studied: catalytic site inhibitors (carbamate insecticides, edrophonium, and benzylpiperidine), peripheral site inhibitors (tubocurarine, ethidium bromide, and propidium iodide), and bivalent inhibitors (donepezil, BW284C51, and a series of bis(n)-tacrines). All were screened on murine AChE to compare and contrast changes of peripheral site conformation in the TFT assay with catalytic inhibition. All the inhibitors reduced thioflavin-T fluorescence in a concentration-dependent manner with potencies (IC50) ranging from 8 nM for bis(6)-tacrine to 159 µM for benzylpiperidine. Potencies in the fluorescence assay were correlated well with their potencies for enzyme inhibition (R2 = 0.884). Efficacies for reducing thioflavin-T fluorescence ranged from 23-36% for catalytic site inhibitors and tubocurarine to near 100% for ethidium bromide and propidium iodide. Maximal efficacies could be reconciled with known mechanisms of interaction of the inhibitors with AChE. When extended to pest species, we anticipate these findings will assist in the discovery and development of novel, selective bivalent insecticides acting on AChE.

16.
J Med Chem ; 56(19): 7615-24, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23984975

ABSTRACT

The molecular interactions between the enzyme acetylcholinesterase (AChE) and two compound classes consisting of N-[2-(diethylamino)ethyl]benzenesulfonamides and N-[2-(diethylamino)ethyl]benzenemethanesulfonamides have been investigated using organic synthesis, enzymatic assays, X-ray crystallography, and thermodynamic profiling. The inhibitors' aromatic properties were varied to establish structure-activity relationships (SAR) between the inhibitors and the peripheral anionic site (PAS) of AChE. The two structurally similar compound classes proved to have distinctly divergent SARs in terms of their inhibition capacity of AChE. Eight X-ray structures revealed that the two sets have different conformations in PAS. Furthermore, thermodynamic profiles of the binding between compounds and AChE revealed class-dependent differences of the entropy/enthalpy contributions to the free energy of binding. Further development of the entropy-favored compound class resulted in the synthesis of the most potent inhibitor and an extension beyond the established SARs. The divergent SARs will be utilized to develop reversible inhibitors of AChE into reactivators of nerve agent-inhibited AChE.


Subject(s)
Acetylcholinesterase/chemistry , Benzene Derivatives/chemistry , Cholinesterase Inhibitors/chemistry , Naphthalenes/chemistry , Sulfonamides/chemistry , Animals , Benzene Derivatives/chemical synthesis , Binding Sites , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cholinesterase Inhibitors/chemical synthesis , Crystallography, X-Ray , Enzyme Assays , Enzyme Reactivators/chemistry , Hydrolysis , Mice , Molecular Structure , Naphthalenes/chemical synthesis , Protein Binding , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Thermodynamics
17.
Biochem Pharmacol ; 85(9): 1389-97, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23376121

ABSTRACT

Nerve agents such as tabun, cyclosarin and Russian VX inhibit the essential enzyme acetylcholinesterase (AChE) by organophosphorylating the catalytic serine residue. Nucleophiles, such as oximes, are used as antidotes as they can reactivate and restore the function of the inhibited enzyme. The oxime HI-6 shows a notably low activity on tabun adducts but can effectively reactivate adducts of cyclosarin and Russian VX. To examine the structural basis for the pronounced substrate specificity of HI-6, we determined the binary crystal structures of Mus musculus AChE (mAChE) conjugated by cyclosarin and Russian VX and found a conformational mobility of the side chains of Phe338 and His447. The interaction between HI-6 and tabun-adducts of AChE were subsequently investigated using a combination of time resolved fluorescence spectroscopy and X-ray crystallography. Our findings show that HI-6 binds to tabun inhibited Homo sapiens AChE (hAChE) with an IC50 value of 300µM and suggest that the reactive nucleophilic moiety of HI-6 is excluded from the phosphorus atom of tabun. We propose that a conformational mobility of the side-chains of Phe338 and His447 is a common feature in nerve-agent adducts of AChE. We also suggest that the conformational mobility allow HI-6 to reactivate conjugates of cyclosarin and Russian VX while a reduced mobility in tabun conjugated AChE results in steric hindrance that prevents efficient reactivation.


Subject(s)
Acetylcholinesterase/chemistry , Antidotes/chemistry , Chemical Warfare Agents/chemistry , Cholinesterase Inhibitors/chemistry , Oximes/chemistry , Pyridinium Compounds/chemistry , Animals , Antidotes/pharmacology , Catalytic Domain , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Organophosphates/chemistry , Organophosphorus Compounds/chemistry , Organothiophosphorus Compounds/chemistry , Oximes/pharmacology , Protein Conformation , Pyridinium Compounds/pharmacology , Spectrometry, Fluorescence , Substrate Specificity
19.
PLoS One ; 6(11): e26039, 2011.
Article in English | MEDLINE | ID: mdl-22140425

ABSTRACT

Acetylcholinesterase (AChE) is an essential enzyme that terminates cholinergic transmission by rapid hydrolysis of the neurotransmitter acetylcholine. Compounds inhibiting this enzyme can be used (inter alia) to treat cholinergic deficiencies (e.g. in Alzheimer's disease), but may also act as dangerous toxins (e.g. nerve agents such as sarin). Treatment of nerve agent poisoning involves use of antidotes, small molecules capable of reactivating AChE. We have screened a collection of organic molecules to assess their ability to inhibit the enzymatic activity of AChE, aiming to find lead compounds for further optimization leading to drugs with increased efficacy and/or decreased side effects. 124 inhibitors were discovered, with considerable chemical diversity regarding size, polarity, flexibility and charge distribution. An extensive structure determination campaign resulted in a set of crystal structures of protein-ligand complexes. Overall, the ligands have substantial interactions with the peripheral anionic site of AChE, and the majority form additional interactions with the catalytic site (CAS). Reproduction of the bioactive conformation of six of the ligands using molecular docking simulations required modification of the default parameter settings of the docking software. The results show that docking-assisted structure-based design of AChE inhibitors is challenging and requires crystallographic support to obtain reliable results, at least with currently available software. The complex formed between C5685 and Mus musculus AChE (C5685•mAChE) is a representative structure for the general binding mode of the determined structures. The CAS binding part of C5685 could not be structurally determined due to a disordered electron density map and the developed docking protocol was used to predict the binding modes of this part of the molecule. We believe that chemical modifications of our discovered inhibitors, biochemical and biophysical characterization, crystallography and computational chemistry provide a route to novel AChE inhibitors and reactivators.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/analysis , Cholinesterase Inhibitors/chemistry , High-Throughput Screening Assays/methods , Models, Molecular , Molecular Targeted Therapy , Acetylcholinesterase/chemistry , Binding Sites , Cholinesterase Inhibitors/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Ligands , Molecular Dynamics Simulation
20.
Biochem Pharmacol ; 80(6): 941-6, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20510679

ABSTRACT

The therapeutic approach of organophosphorus compound (OP) intoxications is to reactivate the inhibited enzyme acetylcholinesterase (AChE). Numerous studies demonstrated a limited efficacy of standard oxime-based reactivators against different nerve agents such as tabun and cyclosarin. This emphasizes research for more effective oximes. In the present study, reactivation kinetics of tabun-, sarin-, cyclosarin-, VX- or paraoxon-ethyl-inhibited human AChE (hAChE) with a homologous series of bis-ortho-pyridiniumaldoximes, Ortho-4 - Ortho-9, was investigated with a robot-assisted setting, allowing determination of second-order reactivation rate constants as well as model calculations. The reactivation constants of Ortho-4 - Ortho-9 resulted in marked differences of affinity and reactivity depending on the OP structure and the linker length of the oximes. In general, the K(D) values decreased with increasing linker length. Reactivity increased from Ortho-4 to Ortho-6 for PXE- and VX-inhibited hAChE and from Ortho-4 to Ortho-7 for GA-inhibited hAChE and decreased again with Ortho-8 and Ortho-9. In contrast, k(r) decreased with increasing linker length for sarin- and cyclosarin-inhibited hAChE. In view of the pronounced decrease of K(D) from Ortho-4 to Ortho-9, the k(r2) values increased with all tested OP. Hence, the ratios of K(I)/K(D) and of K(I)/k(r2) showed that in almost all cases the affinity of Ortho-N to the native hAChE was higher than to OP-inhibited enzyme. Model calculations indicated that Ortho-6 - Ortho-9 could be superior to obidoxime in reactivating tabun-inhibited hAChE. Finally, these data emphasize the need to develop oximes with a higher selective affinity towards OP-inhibited hAChE in order to minimize possible side effects.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Organophosphorus Compounds/pharmacokinetics , Oximes/antagonists & inhibitors , Oximes/metabolism , Pyridinium Compounds/antagonists & inhibitors , Pyridinium Compounds/metabolism , Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Humans , Kinetics , Organophosphorus Compounds/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...