Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Biol Interact ; 259(Pt B): 187-204, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27129421

ABSTRACT

The antidotal treatment of organophosphates (OP) nerve agents (NA) poisoning is based on anticholinergics (e.g. atropine) combined with oxime reactivators (e.g. 2PAM) of acetylcholinesterase (AChE). This treatment is symptomatic and does not degrade the OP. New small-molecule OP scavengers were developed as bifunctional hybrids. Their molecular design was based on combining a nucleophile that directly degrades OP with a moiety that reactivates OP-inhibited AChE. The OP degrading moiety is either benzhydroxamic acid (BHA) or 4-pyridinehydroxamic acid (4PHA) coupled via (CH2)n, (n = 1 or 3) to 2PAM. Three newly synthesized oxime-hydroxamate hybrids: 2PAMPr4PHA, 2PAMMeBHA and 2,4-DiPAMMeBHA were found to detoxify sarin, cyclosarin and soman in solution at 3-10-fold faster rate than 2PAM and to reactivate OP-AChE in vitro. 2PAMPr4PHA displayed 18-fold faster reactivation than 2-PAM of cyclosarin-inhibited HuAChE (kr = 3.6 × 102 vs. 0.2 × 102 M-1min-1, respectively, 37 °C). These hybrids inhibited AChE reversibly, IC50 = 16-48 µM, thereby decreasing the inhibition rates by OPs. The LD50 (im) of 2PAMPr4PHA, 2PAMMeBHA and 2,4DiPAMMeBHA are >568, 508 and >506 µmol/kg in rats and 144, 203 and >506 µmol/kg in guinea pigs. The rate of blood ChE recovery by the hybrids administered either pre- or post-exposure to 0.8xLD50 sarin was comparable or faster than 2PAM. Antidotal efficacy of 2PAMPr4PHA, 2PAMMeBHA and 2,4DiPAMMeBHA administered with atropine, as pre-treatment to sarin in rats (im), yielded protection ratios (PR) 11.6, 11.5 and 4.7, respectively, vs. 5.5 with 2PAM. Post-treatment against various OPs in rats and guinea-pigs yielded PRs higher or similar to that of 2 PAM. Our in vivo data indicates that some hybrids may serve as efficient small molecule scavengers for mitigating the toxicity of OP NAs.


Subject(s)
Nerve Agents/toxicity , Neurotoxins/toxicity , Small Molecule Libraries/pharmacology , Acetylcholinesterase/blood , Acetylcholinesterase/metabolism , Animals , Antidotes/pharmacology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Reactivators/pharmacology , Guinea Pigs , Humans , Hydroxamic Acids/chemistry , Inhibitory Concentration 50 , Kinetics , Male , Organophosphates/toxicity , Organophosphorus Compounds/toxicity , Organothiophosphorus Compounds/toxicity , Oximes/chemistry , Phosphorylation/drug effects , Rats, Sprague-Dawley , Sarin/poisoning , Time Factors , Toxicity Tests, Acute
2.
Neurodegener Dis ; 16(1-2): 95-110, 2016.
Article in English | MEDLINE | ID: mdl-26606130

ABSTRACT

We previously developed orthosteric M1 muscarinic agonists (e.g. AF102B, AF267B and AF292), which act as cognitive enhancers and potential disease modifiers. We now report on a novel compound, AF710B, a highly potent and selective allosteric M1 muscarinic and σ1 receptor agonist. AF710B exhibits an allosteric agonistic profile on the M1 muscarinic receptor; very low concentrations of AF710B significantly potentiated the binding and efficacy of carbachol on M1 receptors and their downstream effects (p-ERK1/2, p-CREB). AF710B (1-30 µg/kg, p.o.) was a potent and safe cognitive enhancer in rats treated with the M1 antagonist trihexyphenidyl (passive avoidance impairment). These effects of AF710B involve σ1 receptor activation. In agreement with its antiamnesic properties, AF710B (at 30 nM), via activation of M1 and a possible involvement of σ1 receptors, rescued mushroom synapse loss in PS1-KI and APP-KI neuronal cultures, while AF267B (1 µM) was less potent in PS1-KI and ineffective in APP-KI models, respectively. In female 3xTg-AD mice, AF710B (10 µg/kg, i.p./daily/2 months) (i) mitigated cognitive impairments in the Morris water maze; (ii) decreased BACE1, GSK3ß activity, p25/CDK5, neuroinflammation, soluble and insoluble Aß40, Aß42, plaques and tau pathologies. AF710B differs from conventional σ1 and M1 muscarinic (orthosteric, allosteric or bitopic) agonists. These results highlight AF710B as a potential treatment for Alzheimer's disease (e.g. improving cognitive deficits, synaptic loss, amyloid and tau pathologies, and neuroinflammation) with a superior profile over a plethora of other therapeutic strategies.


Subject(s)
Alzheimer Disease/drug therapy , Nootropic Agents/pharmacology , Receptor, Muscarinic M1/agonists , Receptors, sigma/agonists , Spiro Compounds/pharmacology , Thiazolidines/pharmacology , Allosteric Regulation , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice, Transgenic , Nootropic Agents/chemistry , PC12 Cells , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Muscarinic M1/metabolism , Receptors, sigma/metabolism , Spiro Compounds/chemistry , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Thiazolidines/chemistry
3.
J Mol Neurosci ; 20(3): 349-56, 2003.
Article in English | MEDLINE | ID: mdl-14501019

ABSTRACT

M1 muscarinic receptors (M1 mAChRs) play a role in an apparent linkage of three major hallmarks of Alzheimer's disease (AD): beta-amyloid (Abeta) peptide; tau hyperphosphorylation and paired helical filaments (PHFs); and loss of cholinergic function conducive to cognitive impairments. We evaluated the M1 muscarinic agonists AF102B (Cevimeline, EVOXAC trade mark : prescribed for Sjøgren's syndrome), AF150(S), and AF267B on some of these hallmarks of AD. Activation of M1 mAChRs with these agonists leads, inter alia, to enhanced secretion of amyloid precursor protein (alpha-APP), (via alpha-secretase activation), to decreased Abeta (via gamma-secretase inhibition), and to inhibition of Abeta- and/or oxidative stress-induced cell death. In several animal models mimicking different aspects of AD, these drugs restored cognitive impairments, and in select cases induced a decrease in brain Abeta elevation, with a high safety margin, following po administration. Notably, in mice with small hippocampi, unlike rivastigmine and nicotine, AF150(S) and AF267B restored cognitive impairments also on escape latency in a Morris water maze paradigm, in reversal learning. Studies from other labs showed that AF102B and talsaclidine (another M1 agonist) decreased cerbrospinal fluid (CSF) Abeta in AD patients following chronic treatment, being the first reported drugs with such a profile. The clinical significance of these studies remains to be elucidated, yet based on in vivo (rabbits) and in vitro studies (cell cultures), our M1 agonists can decrease brain Abeta, owing to a novel and dual complementary effect (e.g., inhibition of gamma-secretase and activation of alpha-secretase). Remarkably, although M1 agonists can decrease CSF Abeta in AD patients, an increased AD-type pathology in Parkinson's disease was recently been associated with chronic antimuscarinic treatment. In another aspect, these agonists decreased tau hyperphosphorylation in vitro and in vivo. Notably, nicotinic agonists or cholinesterase inhibitors increased tau hyperphosphorylation. In summary, the M1 agonists tested are effective on cognition and behavior and show unique disease-modifying properties owing to beneficial effects on major hallmarks of AD. This may place such drugs in the first line of modern AD therapies (e.g., beta- or gamma-secretase inhibitors, vaccines against Abeta, statins, and inhibitors of tau hyperphosphorylation).


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Muscarinic Agonists/pharmacology , Receptor, Muscarinic M1/drug effects , Thiophenes , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/drug effects , Animals , Brain/metabolism , Brain/physiopathology , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Humans , Memory Disorders/drug therapy , Mice , Oxidative Stress/drug effects , Oxidative Stress/physiology , Piperidines/pharmacology , Quinuclidines/pharmacology , Receptor, Muscarinic M1/metabolism , Spiro Compounds/pharmacology , Thiazoles/pharmacology , tau Proteins/drug effects , tau Proteins/metabolism
4.
J Org Chem ; 61(20): 7116-7124, 1996 Oct 04.
Article in English | MEDLINE | ID: mdl-11667614

ABSTRACT

Thiol-mediated free radical isomerization of a deliberately substituted but-3-enyl isocyanide 12a, and n-Bu(3)SnH/AIBN-mediated free radical cyclization of a deliberately substituted but-3-enyl isothiocyanate 22, afforded, respectively, the (ethylthio)pyrroline 13a and the thiopyroglutamates 5 and 23. Reduction, protection, and deprotection of these heterocyclic compounds afforded proline derivatives 6 and 25 which contain all the structural elements of alpha-kainic acid (1) except the C-2 acetic acid moiety. These intermediates were stereospecifically converted into (+/-)-alpha-kainic acid using a new method of temporary sulfur connection. Accordingly, CH(2)CO(2)Me is linked to the chiral isopropenyl anchor and then intramolecularly connected to the pyrrolidine ring and eventually disconnected from its anchor by a sequential reductive double elimination process in which the isopropenyl double bond is restored.

SELECTION OF CITATIONS
SEARCH DETAIL