Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
CNS Neurosci Ther ; 30(6): e14814, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887858

ABSTRACT

AIMS: Alzheimer's disease (AD) is a devastating dementia characterized by extracellular amyloid-ß (Aß) protein aggregates and intracellular tau protein deposition. Clinically available drugs mainly target acetylcholinesterase (AChE) and indirectly sustain cholinergic neuronal tonus. Butyrylcholinesterase (BChE) also controls acetylcholine (ACh) turnover and is involved in the formation of Aß aggregates and senile plaques. UW-MD-95 is a novel carbamate-based compound acting as a potent pseudo-irreversible BChE inhibitor, with high selectivity versus AChE, and showing promising protective potentials in AD. METHODS: We characterized the neuroprotective activity of UW-MD-95 in mice treated intracerebroventricularly with oligomerized Aß25-35 peptide using behavioral, biochemical, and immunohistochemical approaches. RESULTS: When injected acutely 30 min before the behavioral tests (spontaneous alternation in the Y-maze, object recognition, or passive avoidance), UW-MD-95 (0.3-3 mg/kg) showed anti-amnesic effects in Aß25-35-treated mice. When injected once a day over 7 days, it prevented Aß25-35-induced memory deficits. This effect was lost in BChE knockout mice. Moreover, the compound prevented Aß25-35-induced oxidative stress (assessed by lipid peroxidation or cytochrome c release), neuroinflammation (IL-6 and TNFα levels or GFAP and IBA1 immunoreactivity) in the hippocampus and cortex, and apoptosis (Bax level). Moreover, UW-MD-95 significantly reduced the increase in soluble Aß1-42 level in the hippocampus induced by Aß25-35. CONCLUSION: UW-MD-95 appeared as a potent neuroprotective compound in the Aß25-35 model of AD, with potentially an impact on Aß1-42 accumulation that could suggest a novel mechanism of neuroprotection.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Butyrylcholinesterase , Cholinesterase Inhibitors , Disease Models, Animal , Neuroprotective Agents , Peptide Fragments , Animals , Neuroprotective Agents/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Mice , Peptide Fragments/toxicity , Male , Cholinesterase Inhibitors/pharmacology , Butyrylcholinesterase/metabolism , Mice, Inbred C57BL , Maze Learning/drug effects , Dose-Response Relationship, Drug , Oxidative Stress/drug effects
2.
Int J Mol Sci ; 25(7)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38612544

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are the main class of ionotropic receptors for the excitatory neurotransmitter glutamate. They play a crucial role in the permeability of Ca2+ ions and excitatory neurotransmission in the brain. Being heteromeric receptors, they are composed of several subunits, including two obligatory GluN1 subunits (eight splice variants) and regulatory GluN2 (GluN2A~D) or GluN3 (GluN3A~B) subunits. Widely distributed in the brain, they regulate other neurotransmission systems and are therefore involved in essential functions such as synaptic transmission, learning and memory, plasticity, and excitotoxicity. The present review will detail the structure, composition, and localization of NMDARs, their role and regulation at the glutamatergic synapse, and their impact on cognitive processes and in neurodegenerative diseases (Alzheimer's, Huntington's, and Parkinson's disease). The pharmacology of different NMDAR antagonists and their therapeutic potentialities will be presented. In particular, a focus will be given on fluoroethylnormemantine (FENM), an investigational drug with very promising development as a neuroprotective agent in Alzheimer's disease, in complement to its reported efficacy as a tomography radiotracer for NMDARs and an anxiolytic drug in post-traumatic stress disorder.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Humans , Neurodegenerative Diseases/drug therapy , Receptors, N-Methyl-D-Aspartate , Alzheimer Disease/drug therapy , Glutamic Acid
3.
Phytother Res ; 38(2): 694-712, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38011416

ABSTRACT

BACKGROUND AND AIM: By using an in vivo phenotypic screening assay in zebrafish, we identified Convolamine, a tropane alkaloid from Convulvus plauricalis, as a positive modulator of the sigma-1 receptor (S1R). The wfs1abKO zebrafish larva, a model of Wolfram syndrome, exhibits an increased visual-motor response due to a mutation in Wolframin, a protein involved in endoplasmic reticulum-mitochondria communication. We previously reported that ligand activating S1R, restored the cellular and behavioral deficits in patient fibroblasts and zebrafish and mouse models. EXPERIMENTAL PROCEDURES: We screened a library of 108 repurposing and natural compounds on zebrafish motor response. KEY RESULTS: One hit, the tropane alkaloid Convolamine, restored normal mobility in wfs1abKO larvae without affecting wfs1abWT controls. They did not bind to the S1R agonist/antagonist binding site nor dissociated S1R from BiP, an S1R activity assay in vitro, but behaved as a positive modulator by shifting the IC50 value of the reference agonist PRE-084 to lower values. Convolamine restored learning in Wfs1∆Exon8 , Dizocilpine-treated, and Aß25-35 -treated mice. These effects were observed at low ~1 mg/kg doses, not shared by Convolvine, the desmethyl metabolite, and blocked by an S1R antagonist. CONCLUSION AND IMPLICATIONS: Convolamine therefore acts as an S1R positive modulator and this pharmacological action is relevant to the traditional use of Shankhpushpi in memory and cognitive protection.


Subject(s)
Alkaloids , Convolvulus , Receptors, sigma , Humans , Mice , Animals , Sigma-1 Receptor , Receptors, sigma/genetics , Receptors, sigma/metabolism , Zebrafish/metabolism , Alkaloids/pharmacology , Cognition
4.
Neuropharmacology ; 242: 109733, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37844867

ABSTRACT

Fluoroethylnormemantine (FENM) is a Memantine derivative with anti-amnesic and neuroprotective activities showed in the Aß25-35 pharmacological mouse model of Alzheimer's disease (AD). As AD is a complex multi-factorial neurodegenerative pathology, combination therapies relying on drugs acting through different pathways, have been suggested to more adequately address neuroprotection. As several agonists of the sigma-1 receptor (S1R), an intracellular chaperone, are presently in phase 2 or 3 clinical trials in neurodegenetrative diseases including AD, we examined the potentialities of S1R drug-based combinations with FENM, or Memantine. Aß25-35-treated mice were treated with S1R agonists (PRE-084, Igmesine, Cutamesine) and/or FENM, or Memantine, during 7 days after intracerebroventricular administration of oligomerized Aß25-35. Mice were then tested for spatial short-term memory on day 8 and non-spatial long-term memory on days 9-10, using the spontaneous alternation or passive avoidance tests, respectively. The FENM or Memantine combination with Donepezil, that non-selectively inhibits acetylcholinesterase and activates S1R, was also tested. The efficacy of combinations using maximal non-active or minimal active doses of S1R agonist or FENM was analyzed using calculations of the combination index, based on simple isobologram representation. Data showed that most of the FENM-based combinations led to synergistic protection against Aß25-35-induced learning deficits, for both long- and short-term memory responses, with a higher efficiency on the latter. Memantine led to synergistic combination in short-term memory but poorly in long-term memory responses, with either PRE-084 or Donepezil. These study showed that drug combinations based on FENM and S1R agonists may lead to highly effective and synergistic protection in AD, particularly on short-term memory.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Receptors, sigma , Mice , Animals , Memantine/pharmacology , Alzheimer Disease/metabolism , Donepezil/therapeutic use , Acetylcholinesterase , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, sigma/metabolism , Sigma-1 Receptor
5.
J Med Chem ; 66(9): 6414-6435, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37127287

ABSTRACT

We present the synthesis and characterization of merged human butyrylcholinesterase (hBChE) inhibitor/cannabinoid receptor 2 (hCB2R) ligands for the treatment of neurodegeneration. In total, 15 benzimidazole carbamates were synthesized and tested for their inhibition of human cholinesterases, also with regard to their pseudoirreversible binding mode and affinity toward both cannabinoid receptors in radioligand binding studies. After evaluation in a calcium mobilization assay as well as a ß-arrestin 2 (ßarr2) recruitment assay, two compounds with balanced activities on both targets were tested for their immunomodulatory effect on microglia activation and regarding their pharmacokinetic properties and blood-brain barrier penetration. Compound 15d, containing a dimethyl carbamate motif, was further evaluated in vivo, showing prevention of Aß25-35-induced learning impairments in a pharmacological mouse model of Alzheimer's disease for both short- and long-term memory responses. Additional combination studies proved a synergic effect of BChE inhibition and CB2R activation in vivo.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Animals , Mice , Humans , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Neuroprotection , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Cholinesterase Inhibitors/chemistry , Receptors, Cannabinoid , Acetylcholinesterase/metabolism , Structure-Activity Relationship
6.
Toxicology ; 480: 153319, 2022 10.
Article in English | MEDLINE | ID: mdl-36100137

ABSTRACT

Fluoronitrile gas (C4F7N, CAS number 42532-60-5) is one of the most promising candidates as insulating and/or breaking medium in high and medium voltage electrical equipment. Besides its promising properties, C4F7N gas is however not devoid of acute toxicity when used pure or in gas mixtures. The toxicity was not extensively analyzed and reported. The aim of the present study was to analyze in mice the consequences of a single exposure to C4F7N gas, at different concentrations and different timepoints after exposure. Male and female Swiss mice were exposed to breathable air or C4F7N gas, at 800 ppmv or 1500 ppmv, for 4 h on day 0. Behavioral tests (spontaneous alternation in the Y-maze and object recognition) were performed on days 1, 7 and 14 to assess memory alterations. The animals were then sacrificed and their brains dissected for biochemical analyses or fixed with paraformaldehyde for histology and immunohistochemistry. Results showed behavioral impairments and memory deficits, with impairments of alternation at days 1 and 7 and object recognition at day 14. Histological alterations of pyramidal neuronal layer in the hippocampus, neuroinflammatory astroglial reaction, and microglial alterations were observed, more marked in female than male mice. Moreover, the biochemical analyses done in the brain of 1500 ppmv exposed female mice showed a reductive stress with decreased lipid peroxidation and release of cytochrome c, leading to apoptosis with increases in caspase-9 cleavage and γ-H2AX/H2AX ratio. Finally, electrophysiological analyses using a multi-electrode array allowed the measure of the extracellular activity of pyramidal neurons in the CA2 area and revealed that exposure to the gas not only prevented the induction of long-term potentiation but even provoked an epileptoid-like activity in some neurons suggesting major alterations of synaptic plasticity. This study therefore showed that an acute exposure of mice to C4F7N gas provoked, particularly in female animals, memory alterations and brain toxicity characterized by a reductive stress, microglial toxicity, loss of synaptic plasticity and apoptosis. Its use in industrial installations must be done with extreme caution.


Subject(s)
Cytochromes c , Neurotoxicity Syndromes , Animals , Brain/pathology , Caspase 9 , Female , Hippocampus/pathology , Male , Memory Disorders/pathology , Mice , Neuronal Plasticity/physiology , Neurotoxicity Syndromes/pathology
7.
J Am Chem Soc ; 144(7): 3279-3284, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138833

ABSTRACT

To develop tools to investigate the biological functions of butyrylcholinesterase (BChE) and the mechanisms by which BChE affects Alzheimer's disease (AD), we synthesized several selective, nanomolar active, pseudoirreversible photoswitchable BChE inhibitors. The compounds were able to specifically influence different kinetic parameters of the inhibition process by light. For one compound, a 10-fold difference in the IC50-values (44.6 nM cis, 424 nM trans) in vitro was translated to an "all or nothing" response with complete recovery in a murine cognition-deficit AD model at dosages as low as 0.3 mg/kg.


Subject(s)
Alzheimer Disease/drug therapy , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/therapeutic use , Cognition/drug effects , Neuroprotective Agents/therapeutic use , Nootropic Agents/therapeutic use , Alzheimer Disease/chemically induced , Amyloid beta-Peptides , Animals , Azo Compounds/chemical synthesis , Azo Compounds/metabolism , Azo Compounds/radiation effects , Azo Compounds/therapeutic use , Carbamates/chemical synthesis , Carbamates/metabolism , Carbamates/radiation effects , Carbamates/therapeutic use , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/radiation effects , Kinetics , Mice , Molecular Docking Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/radiation effects , Nootropic Agents/chemical synthesis , Nootropic Agents/metabolism , Nootropic Agents/radiation effects , Peptide Fragments , Protein Binding , Stereoisomerism
8.
Neuropharmacology ; 186: 108467, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33516737

ABSTRACT

Huntington's disease (HD) is due to a mutation in the gene encoding for Huntingtin protein generating polyQ domain extension. Mutant Htt (mHtt) leads to important dysfunction of the BDNF/TrkB signaling. We previously described the 23aa Htt fragment P42, that attenuated the pathological phenotypes induced by mHtt. We reported that, in the R6/2 mouse model of HD, P42 rescued striatal TrkB level but marginally increased cortical BDNF. In the present study, our aim was to address P42 neuroprotection in presence of an external input of BDNF. We combined P42 administration with environmental enrichment (EE), induced by training in the Hamlet test. We examined the consequences of P42 + EE combination on different phenotypes in R6/2 HD mice: motor and cognitive performances, recorded at early and late pathological stages, and analyzed aggregated mHtt and BDNF levels in forebrain structures. Hamlet exploration (i.e., entries in Run, Hide, Eat, Drink and Interact houses) was gradually impaired in R6/2 mice, but maintained by P42 treatment until week 8. Topographic memory alteration measured at week 7 was attenuated by P42. Motor performances (rotarod) were significantly ameliorated by the P42 + EE combination until late stage (week 12). The P42 + EE combination also significantly decreased aggregated Htt levels in the hippocampus, striatum and cortex, and increased BDNF levels in the cortex and striatum. We concluded that combination between P42 treatment, known to increase TrkB striatal expression, and a BDNF-enhancing therapy such as EE efficiently delayed HD pathology in R6/2 mice. Use of dual therapies might be a pertinent strategy to fight neurodegeneration in HD.


Subject(s)
Environment , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Peptide Fragments/administration & dosage , Amino Acid Sequence , Animals , Combined Modality Therapy/methods , Female , Huntingtin Protein/genetics , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Peptide Fragments/genetics , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...