Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Chembiochem ; 23(23): e202200456, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36193860

ABSTRACT

We report the implementation of coordination complexes containing two types of cationic moieties, i. e. pyridinium and ammonium quaternary salt, as potential inhibitors of human cholinesterase enzymes. Utilization of ligands containing NNO-coordination site and binding zinc metal ion allowed mono- and tetra-nuclear complexes to be obtained with corner and grid structural type, respectively, thus affecting the overall charge of the compounds (from +1 to +8). We were able to examine for the first time the multivalency effect of metallosupramolecular species on their inhibitory abilities towards acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Importantly, resolution of the crystal structures of the obtained enzyme-substrate complexes provided a better understanding of the inhibition process at the molecular level.


Subject(s)
Acetylcholinesterase , Butyrylcholinesterase , Humans , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Ligands , Cations , Molecular Docking Simulation
2.
Mol Biol Rep ; 48(10): 6729-6738, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34436724

ABSTRACT

BACKGROUND: Basal stem/progenitor cells of airway epithelium from chronic obstructive pulmonary disease (COPD) patients have a decrease in differentiation and self-renewal potential. Our study aimed at identifying deregulations in the genetic program of these cells that could account for their exhaustion, focusing on genes downstream of the epithelial-mesenchymal transition-inducing transcription factor Slug/Snail2 and responding to transforming growth factor (TGF)-ß. TGF-ß is at higher levels in COPD patient lungs, plays a role in stem/progenitor cell fate and regulates the expression of Slug/Snail2 that is highly expressed in airway basal stem/progenitors. METHODS AND RESULTS: We reanalyzed a gene expression dataset that we generated from COPD and normal primary bronchial basal progenitor cells knocked down for Slug/Snail2 gene. Among the genes that we identified to be repressed downstream of Slug/Snail2 in COPD, we selected those responding to differentiation and TGF-ß. The large majority of these genes are upregulated with differentiation but repressed by TGF-ß. Pathway and ontology enrichment analysis revealed a set of genes coding for transcription factors involved in stem cell maintenance that are repressed downstream of Slug/Snail2 and by TGF-ß in COPD but not normal basal progenitor cells. We also reveal a link between Slug/Snail2 expression and the repressive effect of TGF-ß on these stem cell maintenance genes. CONCLUSION: Our work brings a new insight and molecular perspective to the exhaustion of basal stem/progenitor cells observed in the airway epithelium of COPD patients, revealing that stem cell maintenance genes are repressed in these cells, with TGF-ß and Slug/Snail2 being involved in this deregulation.


Subject(s)
Bronchi/pathology , Epithelium/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Snail Family Transcription Factors/metabolism , Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Female , Humans , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism
3.
Arch Toxicol ; 95(3): 985-1001, 2021 03.
Article in English | MEDLINE | ID: mdl-33517499

ABSTRACT

To date, the only treatments developed for poisoning by organophosphorus compounds, the most toxic chemical weapons of mass destruction, have exhibited limited efficacy and versatility. The available causal antidotes are based on reactivation of the enzyme acetylcholinesterase (AChE), which is rapidly and pseudo-irreversibly inhibited by these agents. In this study, we developed a novel series of monoquaternary reactivators combining permanently charged moieties tethered to position 6- of 3-hydroxypyridine-2-aldoxime reactivating subunit. Highlighted representatives (21, 24, and 27; also coded as K1371, K1374, and K1375, respectively) that contained 1-phenylisoquinolinium, 7-amino-1-phenylisoquinolinium and 4-carbamoylpyridinium moieties as peripheral anionic site ligands, respectively, showed efficacy superior or comparable to that of the clinically used standards. More importantly, these reactivators exhibited wide-spectrum efficacy and were minutely investigated via determination of their reactivation kinetics in parallel with molecular dynamics simulations to study their mechanisms of reactivation of the tabun-inhibited AChE conjugate. To further confirm the potential applicability of these candidates, a mouse in vivo assay was conducted. While K1375 had the lowest acute toxicity and the most suitable pharmacokinetic profile, the oxime K1374 with delayed elimination half-life was the most effective in ameliorating the signs of tabun toxicity. Moreover, both in vitro and in vivo, the versatility of the agents was substantially superior to that of clinically used standards. Their high efficacy and broad-spectrum capability make K1374 and K1375 promising candidates that should be further investigated for their potential as nerve agents and insecticide antidotes.


Subject(s)
Acetylcholinesterase/drug effects , Antidotes/pharmacology , Cholinesterase Reactivators/pharmacology , Acetylcholinesterase/metabolism , Animals , Antidotes/chemical synthesis , Antidotes/chemistry , Cholinesterase Reactivators/chemical synthesis , Cholinesterase Reactivators/chemistry , Female , Male , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , Oximes/chemical synthesis , Oximes/chemistry , Oximes/pharmacology , Structure-Activity Relationship
4.
Chemistry ; 26(65): 15035-15044, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-32633095

ABSTRACT

Novel 6-alkyl- and 6-alkenyl-3-fluoro-2-pyridinaldoximes have been synthesised by using a mild and efficient chemoselective hydrogenation of 6-alkynyl-3-fluoro-2-pyridinaldoxime scaffolds, without altering the reducible, unprotected, sensitive oxime functionality and the C-F bond. These novel 6-alkyl-3-fluoro-2-pyridinaldoximes may find medicinal application as antidotes to organophosphate poisoning. Indeed, one low-molecular-weight compound exhibited increased affinity for sarin-inhibited acetylcholinesterase (hAChE) and greater reactivation efficiency or resurrection for sarin-inhibited hAChE, compared with those of 2-pyridinaldoxime (2-PAM) and 1-({[4-(aminocarbonyl)pyridinio]methoxy}methyl)-2-[(hydroxyimino)methyl]pyridinium chloride (HI-6), two pyridinium salts currently used as antidote by several countries. In addition, the uncharged 3-fluorinated bifunctional hybrid showed increased in vitro blood-brain barrier permeability compared with those of 2-PAM, HI-6 and obidoxime. These promising features of novel low-molecular-weight alkylfluoropyridinaldoxime open up a new era for the design, synthesis and discovery of central non-quaternary broad spectrum reactivators for organophosphate-inhibited cholinesterases.


Subject(s)
Blood-Brain Barrier , Acetylcholinesterase/metabolism , Blood-Brain Barrier/metabolism , Cholinesterase Inhibitors , Cholinesterase Reactivators , Humans , Hydrogenation , Oximes , Permeability , Pyridinium Compounds , Sarin
5.
Bioorg Med Chem Lett ; 30(24): 127609, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33039562

ABSTRACT

Acetylcholinesterase inhibitors are the mainstay of Alzheimer's disease treatments, despite having only short-term symptomatic benefits and severe side effects. Selective butyrylcholinesterase inhibitors (BuChEIs) may be more effective treatments in late-stage Alzheimer's disease with fewer side effects. Virtual screening is a powerful tool for identifying potential inhibitors in large digital compound databases. This study used structure-based virtual screening combined with physicochemical filtering to screen the InterBioScreen and Maybridge databases for novel selective BuChEIs. The workflow rapidly identified 22 potential hits in silico, resulting in the discovery of a human BuChEI with low-micromolar potency in vitro (IC50 2.4 µM) and high selectivity for butyrylcholinesterase over acetylcholinesterase. The compound was a rapidly reversible BuChEI with mixed-model in vitro inhibition kinetics. The binding interactions were investigated using in silico molecular dynamics and by developing structure-activity relationships using nine analogues. The compound also displayed high permeability in an in vitro model of the blood-brain barrier.


Subject(s)
Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Drug Discovery , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Drug Design , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Structure-Activity Relationship
6.
J Med Chem ; 65(6): 4649-4666, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35255209

ABSTRACT

Recent events demonstrated that organophosphorus nerve agents are a serious threat for civilian and military populations. The current therapy includes a pyridinium aldoxime reactivator to restore the enzymatic activity of acetylcholinesterase located in the central nervous system and neuro-muscular junctions. One major drawback of these charged acetylcholinesterase reactivators is their poor ability to cross the blood-brain barrier. In this study, we propose to evaluate glucoconjugated oximes devoid of permanent charge as potential central nervous system reactivators. We determined their in vitro reactivation efficacy on inhibited human acetylcholinesterase, the crystal structure of two compounds in complex with the enzyme, their protective index on intoxicated mice, and their pharmacokinetics. We then evaluated their endothelial permeability coefficients with a human in vitro model. This study shed light on the structural restrains of new sugar oximes designed to reach the central nervous system through the glucose transporter located at the blood-brain barrier.


Subject(s)
Organophosphate Poisoning , Acetylcholinesterase , Animals , Antidotes/pharmacology , Antidotes/therapeutic use , Cholinesterase Inhibitors/pharmacology , Mice , Organophosphate Poisoning/drug therapy , Organophosphorus Compounds/pharmacology , Oximes/chemistry , Oximes/pharmacology , Oximes/therapeutic use , Sugars
7.
Stem Cell Rev Rep ; 17(3): 703-718, 2021 06.
Article in English | MEDLINE | ID: mdl-33495975

ABSTRACT

Slug/Snail2 belongs to the Epithelial-Mesenchymal Transition (EMT)-inducing transcription factors involved in development and diseases. Slug is expressed in adult stem/progenitor cells of several epithelia, making it unique among these transcription factors. To investigate Slug role in human bronchial epithelium progenitors, we studied primary bronchial basal/progenitor cells in an air-liquid interface culture system that allows regenerating a bronchial epithelium. To identify Slug downstream genes we knocked down Slug in basal/progenitor cells from normal subjects and subjects with COPD, a respiratory disease presenting anomalies in the bronchial epithelium and high levels of TGF-ß in the lungs. We show that normal and COPD bronchial basal/progenitors, even when treated with TGF-ß, express both epithelial and mesenchymal markers, and that the epithelial marker E-cadherin is not a target of Slug and, moreover, positively correlates with Slug. We reveal that Slug downstream genes responding to both differentiation and TGF-ß are different in normal and COPD progenitors, with in particular a set of proliferation-related genes that are among the genes repressed downstream of Slug in normal but not COPD. In COPD progenitors at the onset of differentiation in presence of TGF-ß,we show that there is positive correlations between the effect of differentiation and TGF-ß on proliferation-related genes and on Slug protein, and that their expression levels are higher than in normal cells. As well, the expression of Smad3 and ß-Catenin, two molecules from TGF-ßsignaling pathways, are higher in COPD progenitors, and our results indicate that proliferation-related genes and Slug protein are increased by different TGF-ß-induced mechanisms.


Subject(s)
Bronchi , Pulmonary Disease, Chronic Obstructive , Snail Family Transcription Factors , Stem Cells , Transforming Growth Factor beta , Adult , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cell Proliferation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
8.
Chem Biol Drug Des ; 97(5): 1048-1058, 2021 05.
Article in English | MEDLINE | ID: mdl-33455074

ABSTRACT

Cholinesterase inhibitors remain the mainstay of Alzheimer's disease treatment, and the search for new inhibitors with better efficacy and side effect profiles is ongoing. Virtual screening (VS) is a powerful technique for searching large compound databases for potential hits. This study used a sequential VS workflow combining ligand-based VS, molecular docking and physicochemical filtering to screen for central nervous system (CNS) drug-like acetylcholinesterase inhibitors (AChEIs) amongst the 6.9 million compounds of the CoCoCo database. Eleven in silico hits were initially selected, resulting in the discovery of an AChEI with a Ki of 3.2 µM. In vitro kinetics and in silico molecular dynamics experiments informed the selection of an additional seven analogues. This led to the discovery of two further AChEIs, with Ki values of 2.9 µM and 0.65 µM. All three compounds exhibited reversible, mixed inhibition of acetylcholinesterase. Importantly, the in silico physicochemical filter facilitated the discovery of CNS drug-like compounds, such that all three inhibitors displayed high in vitro blood-brain barrier model permeability.


Subject(s)
Acetylcholinesterase/chemistry , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Binding Sites , Butyrylcholinesterase/metabolism , Catalytic Domain , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Databases, Chemical , Donepezil/chemistry , Donepezil/metabolism , Donepezil/therapeutic use , Electrophorus/metabolism , Horses/metabolism , Kinetics , Molecular Dynamics Simulation , Permeability/drug effects
9.
Biomolecules ; 10(6)2020 06 04.
Article in English | MEDLINE | ID: mdl-32512884

ABSTRACT

(1) Background: Human exposure to organophosphorus compounds employed as pesticides or as chemical warfare agents induces deleterious effects due to cholinesterase inhibition. One therapeutic approach is the reactivation of inhibited acetylcholinesterase by oximes. While currently available oximes are unable to reach the central nervous system to reactivate cholinesterases or to display a wide spectrum of action against the variety of organophosphorus compounds, we aim to identify new reactivators without such drawbacks. (2) Methods: This study gathers an exhaustive work to assess in vitro and in vivo efficacy, and toxicity of a hybrid tetrahydroacridine pyridinaldoxime reactivator, KM297, compared to pralidoxime. (3) Results: Blood-brain barrier crossing assay carried out on a human in vitro model established that KM297 has an endothelial permeability coefficient twice that of pralidoxime. It also presents higher cytotoxicity, particularly on bone marrow-derived cells. Its strong cholinesterase inhibition potency seems to be correlated to its low protective efficacy in mice exposed to paraoxon. Ventilatory monitoring of KM297-treated mice by double-chamber plethysmography shows toxic effects at the selected therapeutic dose. This breathing assessment could help define the No Observed Adverse Effect Level (NOAEL) dose of new oximes which would have a maximum therapeutic effect without any toxic side effects.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Pralidoxime Compounds/pharmacology , Animals , Blood-Brain Barrier/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Injections, Intraperitoneal , Male , Mice , Molecular Structure , Pralidoxime Compounds/chemistry , Recombinant Proteins/metabolism
10.
Toxicology ; 411: 15-23, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30359675

ABSTRACT

The efficiency of human butyrylcholinesterase (BChE) as a stoichiometric bioscavenger of nerve agents is well established. However, wide use is currently limited by production and purification costs. Aiming at identifying an alternative human protein bioscavenger, we looked for an original scaffold candidate by virtual screening of the Protein Data Bank for functional similarity using the "Surfing the Molecules" software (sumo-pbil.ibcp.fr) and a search model based on the BChE active site topology. Besides the expected acetylcholinesterase and butyrylcholinesterase, we identified a set of bile salt activated lipases structures, among which the human pancreatic lipase (hBAL) that shares 34% identity with BChE. We produced the recombinant enzyme in mammalian cells, purified it, and measured the inhibition constants for paraoxon and surrogates of VX, sarin and tabun. We solved the X-ray structure of apo hBAL and conjugates with paraoxon and the surrogates at resolutions in the 2-Å range. These structures allow the assessment of hBAL for scavenging nerve agents. They revealed that hBAL has inverted stereoselectivity for the surrogates of nerve agent compared to human cholinesterases. We observed a remarkable flip of the catalytic histidine driven by the chelation of Zn2+. Dealkylation of the conjugate, aka aging, was solely observed for paraoxon.


Subject(s)
Lipase/chemistry , Nerve Agents/chemistry , Nerve Agents/toxicity , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Catalysis , Chelating Agents/chemistry , Cholinesterase Inhibitors/toxicity , Computer Simulation , Crystallography, X-Ray , Histidine/chemistry , Humans , Kinetics , Lipase/drug effects , Models, Molecular , Pancreas/drug effects , Pancreas/enzymology , Paraoxon/toxicity , Stereoisomerism , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL