Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biol Psychiatry ; 90(7): 458-472, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34274107

ABSTRACT

BACKGROUND: Major depressive disorder is a common, recurrent illness. Recent studies have implicated the NMDA receptor in the pathophysiology of major depressive disorder. (R,S)-ketamine, an NMDA receptor antagonist, is an effective antidepressant but has numerous side effects. Here, we characterized a novel NMDA receptor antagonist, fluoroethylnormemantine (FENM), to determine its effectiveness as a prophylactic and/or antidepressant against stress-induced maladaptive behavior. METHODS: Saline, memantine (10 mg/kg), (R,S)-ketamine (30 mg/kg), or FENM (10, 20, or 30 mg/kg) was administered before or after contextual fear conditioning in 129S6/SvEv mice. Drug efficacy was assayed using various behavioral tests. Protein expression in the hippocampus was quantified with immunohistochemistry or Western blotting. In vitro radioligand binding was used to assay drug binding affinity. Patch clamp electrophysiology was used to determine the effect of drug administration on glutamatergic activity in ventral hippocampal cornu ammonis 3 (vCA3) 1 week after injection. RESULTS: Given after stress, FENM decreased behavioral despair and reduced perseverative behavior. When administered after re-exposure, FENM facilitated extinction learning. As a prophylactic, FENM attenuated learned fear and decreased stress-induced behavioral despair. FENM was behaviorally effective in both male and female mice. (R,S)-ketamine, but not FENM, increased expression of c-fos in vCA3. Both (R,S)-ketamine and FENM attenuated large-amplitude AMPA receptor-mediated bursts in vCA3, indicating a common neurobiological mechanism for further study. CONCLUSIONS: Our results indicate that FENM is a novel drug that is efficacious when administered at various times before or after stress. Future work will further characterize FENM's mechanism of action with the goal of clinical development.


Subject(s)
Depressive Disorder, Major , Ketamine , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Female , Ketamine/pharmacology , Male , Memantine/analogs & derivatives , Mice , Stress, Psychological
2.
Int J Neuropsychopharmacol ; 24(6): 519-531, 2021 07 14.
Article in English | MEDLINE | ID: mdl-33631001

ABSTRACT

BACKGROUND: Memantine, a noncompetitive N-methyl-D-aspartate receptor antagonist, has been approved for use in Alzheimer's disease, but an increasing number of studies have investigated its utility for neuropsychiatric disorders. Here, we characterized a novel compound, fluoroethylnormemtantine (FENM), which was derived from memantine with an extra Fluor in an optimized position for in vivo biomarker labeling. We sought to determine if FENM produced similar behavioral effects as memantine and/or if FENM has beneficial effects against fear, avoidance, and behavioral despair. METHODS: We administered saline, FENM, or memantine prior to a number of behavioral assays, including paired-pulse inhibition, open field, light dark test, forced swim test, and cued fear conditioning in male Wistar rats. RESULTS: Unlike memantine, FENM did not produce nonspecific side effects and did not alter sensorimotor gating or locomotion. FENM decreased immobility in the forced swim test. Moreover, FENM robustly facilitated fear extinction learning when administered prior to either cued fear conditioning training or tone reexposure. CONCLUSIONS: These results suggest that FENM is a promising, novel compound that robustly reduces fear behavior and may be useful for further preclinical testing.


Subject(s)
Behavior, Animal/physiology , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Memantine/analogs & derivatives , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Male , Rats, Wistar
3.
Int J Neuropsychopharmacol ; 24(2): 142-157, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32977336

ABSTRACT

BACKGROUND: Current therapies in Alzheimer's disease (AD), including Memantine, have proven to be only symptomatic but not curative or disease modifying. Fluoroethylnormemantine (FENM) is a structural analogue of Memantine, functionalized with a fluorine group that allowed its use as a positron emission tomography tracer. We here analyzed FENM neuroprotective potential in a pharmacological model of AD compared with Memantine. METHODS: Swiss mice were treated intracerebroventricularly with aggregated Aß 25-35 peptide and examined after 1 week in a battery of memory tests (spontaneous alternation, passive avoidance, object recognition, place learning in the water-maze, topographic memory in the Hamlet). Toxicity induced in the mouse hippocampus or cortex was analyzed biochemically or morphologically. RESULTS: Both Memantine and FENM showed symptomatic anti-amnesic effects in Aß 25-35-treated mice. Interestingly, FENM was not amnesic when tested alone at 10 mg/kg, contrarily to Memantine. Drugs injected once per day prevented Aß 25-35-induced memory deficits, oxidative stress (lipid peroxidation, cytochrome c release), inflammation (interleukin-6, tumor necrosis factor-α increases; glial fibrillary acidic protein and Iba1 immunoreactivity in the hippocampus and cortex), and apoptosis and cell loss (Bcl-2-associated X/B-cell lymphoma 2 ratio; cell loss in the hippocampus CA1 area). However, FENM effects were more robust than observed with Memantine, with significant attenuations vs the Aß 25-35-treated group. CONCLUSIONS: FENM therefore appeared as a potent neuroprotective drug in an AD model, with a superior efficacy compared with Memantine and an absence of direct amnesic effect at higher doses. These results open the possibility to use the compound at more relevant dosages than those actually proposed in Memantine treatment for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amnesia/drug therapy , Memantine/analogs & derivatives , Memantine/pharmacology , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/prevention & control , Amnesia/chemically induced , Amnesia/prevention & control , Amyloid beta-Peptides/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Male , Memantine/administration & dosage , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Peptide Fragments/pharmacology
4.
Amino Acids ; 52(11-12): 1559-1580, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33191465

ABSTRACT

Drug compounds including memantine moieties are an important group of biologically active agents for different pathologies, including the Alzheimer's disease. In the present study, a series of memantine derivatives incorporating amino acid residues have been synthesized and their neuroprotective in vitro evaluation in respect of the Alzheimer's disease, involving the effects on the resistance to Aß toxicity, excitotoxicity, oxidative stress, hypoxia, and neuroinflammation has been studied. The cytotoxicities of the compounds were detected by CPE assay. TC50 and IC50 were determined using Reed and Muench method. Solubility and distribution were measured using a shake-flask method. Permeability of the compounds was studied using Franz diffusion cell and Permeapad™ barrier. These compounds displayed apparent multi-neuroprotective effects against copper-triggered Aß toxicity, glutamate-induced excitotoxicity, and oxidative and hypoxic injuries. They also showed the ability to inhibit the inflammatory cytokine release from the activated microglia and potential anti-neuroinflammatory effects. Especially, two most promising compounds H-4-F-Phe-memantine and H-Tyr-memantine demonstrated the equivalent functional bioactivities in comparison with the positive control memantine hydrochloride. Higher solubility in muriatic buffer than in phosphate buffer was detected. The distribution coefficients showed the optimal lipophilicity for compounds. The presented results propose new class of memantine derivatives as potential drug compounds. Based on the experimental results, the correlations have been obtained between the biological, physicochemical parameters and structural descriptors. The correlation equations have been proposed to predict the properties of new memantine derivatives knowing only the structural formula.


Subject(s)
Alzheimer Disease/drug therapy , Influenza, Human/drug therapy , Memantine/pharmacology , Neuroprotective Agents/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/toxicity , Animals , Dogs , Glutamic Acid/metabolism , Humans , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Memantine/analogs & derivatives , Memantine/chemistry , Neuroprotective Agents/chemistry , Orthomyxoviridae/drug effects , Orthomyxoviridae/pathogenicity , Oxidative Stress/drug effects
5.
Eur J Med Chem ; 181: 111572, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31404859

ABSTRACT

The discovery and development of multitarget-directed ligands (MTDLs) is a promising strategy to find new therapeutic solutions for neurodegenerative diseases (NDs), in particular for Alzheimer's disease (AD). Currently approved drugs for the clinical management of AD are based on a single-target strategy and focus on restoring neurotransmitter homeostasis. Finding disease-modifying therapies AD and other NDs remains an urgent unmet clinical need. The growing consensus that AD is a multifactorial disease, with several interconnected and deregulated pathological pathways, boosted an intensive research in the design of MTDLs. Due to this scientific boom, the knowledge behind the development of MTDLs remains diffuse and lacks balanced guidelines. To rationalize the large amount of data obtained in this field, we herein revise the progress made over the last 5 years on the development of MTDLs inspired by drugs approved for AD. Due to their putative therapeutic benefit in AD, MTDLs based on MAO-B inhibitors will also be discussed in this review.


Subject(s)
Alzheimer Disease/drug therapy , Chemistry Techniques, Synthetic , Drug Design , Drug Discovery , Animals , Chemistry Techniques, Synthetic/methods , Donepezil/analogs & derivatives , Donepezil/chemical synthesis , Donepezil/pharmacology , Dopamine Agents/chemical synthesis , Dopamine Agents/chemistry , Dopamine Agents/pharmacology , Drug Discovery/methods , Humans , Indans/chemical synthesis , Indans/chemistry , Indans/pharmacology , Memantine/analogs & derivatives , Memantine/chemical synthesis , Memantine/pharmacology , Molecular Targeted Therapy , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Nootropic Agents/chemical synthesis , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Rivastigmine/analogs & derivatives , Rivastigmine/chemical synthesis , Rivastigmine/pharmacology , Tacrine/analogs & derivatives , Tacrine/chemical synthesis , Tacrine/pharmacology
6.
Sci Rep ; 9(1): 4612, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874573

ABSTRACT

Hydrogen sulphide has recently drawn much attention due to its potent anti-inflammatory and neuroprotective roles in brain functions. The purpose of the current study was to exploit these beneficial properties of H2S to design a new agent for the treatment of Alzheimer's disease (AD). To pursue our aims, we replaced the free amine group of memantine with an isothiocyanate functionality as a putative H2S-donor moiety. The new chemical entity, named memit, was then tested in vitro to determine whether it retains the pharmacological profile of the "native drug", while also providing a source of H2S in the CNS. Indeed, Memit showed the ability to release H2S through a cysteine-mediated mechanism, thus generating memantine. Moreover, the new hybrid molecule exerts protective effects against neuronal inflammation and induces a drastic fall in ROS production. In addition, memit was also able to reduce the Aß(1-42) self-induced aggregation and exerted cytoprotective effect against Aß oligomers-induced damage in both human neurons and rat microglia cells. Finally, similarly to memantine, the new compound promotes autophagy, a complex process required for cellular homeostasis in cell survival that results to be altered in neurodegenerative diseases. In conclusion, our study revealed that memit is a prodrug of memantine. Further in vivo studies will be necessary to fully investigate the synergic or cumulative effects due to the H2S-releasing moiety and the native drug.


Subject(s)
Alzheimer Disease/drug therapy , Memantine/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Autophagy/drug effects , Cell Line , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Inflammation , Memantine/analogs & derivatives , Memantine/metabolism , Microglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Primary Cell Culture , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species
7.
Nucl Med Biol ; 59: 1-8, 2018 04.
Article in English | MEDLINE | ID: mdl-29413751

ABSTRACT

INTRODUCTION: The aim of this work was to study the biodistribution, metabolism and radiation dosimetry of rats injected with [18F]FNM using PET/CT images. This novel radiotracer targeting NMDA receptor has potential for investigation for neurological and psychiatric diseases. METHODS: Free fraction and stability in fresh human plasma were determined in vitro. PET/CT was performed on anesthetized rats. Organs were identified and 3D volumes of interest (VOIs) were manually drawn on the CT in the center of each organ. Time activity curves (TACs) were created with these VOIs, enabling the calculation of residence times. To confirm these values, ex vivo measurements of organs were performed. Plasma and urine were also collected to study in vivo metabolism. Data was extrapolated to humans, effective doses were estimated using ICRP-60 and ICRP-89 dosimetric models and absorbed doses were estimated using OLINDA/EXM V1.0 and OLINDA/EXM V2.0 (which use weighting factors from ICRP-103 to do the calculations). RESULTS: The [18F]FNM was stable in human plasma and the diffusible free fraction was 53%. As with memantine, this tracer is poorly metabolized in vivo. Ex vivo distributions validated PET/CT data as well as demonstrating a decrease of radiotracer uptake in the brain due to anesthesia. Total effective dose was around 6.11 µSv/MBq and 4.65 µSv/MBq for female and male human dosimetric models, respectively. CONCLUSIONS: This study shows that the presented compound exhibits stability in plasma and plasma protein binding very similar to memantine. Its dosimetry shows that it is suitable for use in humans due to a low total effective dose compared to other PET radiotracers.


Subject(s)
Memantine/analogs & derivatives , Positron Emission Tomography Computed Tomography , Whole Body Imaging , Animals , Blood Proteins/metabolism , Drug Stability , Female , Humans , Memantine/chemical synthesis , Memantine/metabolism , Memantine/pharmacokinetics , Radiometry , Rats , Rats, Sprague-Dawley , Tissue Distribution
8.
Bioorg Med Chem Lett ; 28(4): 689-693, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29366650

ABSTRACT

Owning to the promising neuroprotective profile and the ability to cross the blood-brain barrier, triptolide has attracted extensive attention. Although its limited solubility and toxicity have greatly hindered clinical translation, triptolide has nonetheless emerged as a promising candidate for structure-activity relationship studies for Alzheimer's disease. In the present study, a series of triptolide analogs were designed and synthesized, and their neuroprotective and anti-neuroinflammatory effects were then tested using a cell culture model. Among the triptolide derivatives tested, a memantine conjugate, compound 8, showed a remarkable neuroprotective effect against Aß1-42 toxicity in primary cortical neuron cultures as well as an inhibitory effect against LPS-induced TNF-α production in BV2 cells at a subnanomolar concentration. Our findings provide insight into the different pharmacophores that are responsible for the multifunctional effects of triptolide in the central nervous system. Our study should help in the development of triptolide-based multifunctional anti-Alzheimer drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diterpenes/pharmacology , Neuroprotective Agents/pharmacology , Phenanthrenes/pharmacology , Amyloid beta-Peptides/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line, Transformed , Diterpenes/chemical synthesis , Diterpenes/chemistry , Epoxy Compounds/chemical synthesis , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Memantine/analogs & derivatives , Memantine/chemical synthesis , Memantine/chemistry , Memantine/pharmacology , Mice , Neurons/drug effects , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Peptide Fragments/adverse effects , Phenanthrenes/chemical synthesis , Phenanthrenes/chemistry , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
Nitric Oxide ; 79: 68-83, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29030124

ABSTRACT

Glioblastoma multiforme (GBM) is a devastating brain cancer with no curative treatment. Targeting Nitric Oxide (NO) and glutamatergic pathways may help as adjunctive treatments in GBM. NO at low doses promotes tumorigenesis, while at higher levels (above 300 nM) triggers apoptosis. Gliomas actively secrete high amounts of glutamate which activates EGR signaling and mediates degradation of peritumoral tissues via excitotoxic injury. Memantine inhibits NMDA-subtype of glutamate receptors (NMDARs) and induces autophagic death of glioma cells in vitro and blocks glioma growth in vivo. Nitro-memantines may exert further benefits by limiting NMDAR signaling and by delivery of NO to the areas of excessive NMDAR activity leading NO-accumulation at tumoricidal levels within gliomas. Due to the duality of NO in tumorigenesis, agents which attenuate NO levels may also act beneficial in treatment of GBM. Nitrone compounds including N-tert-Butyl-α-phenylnitrone (PBN) and its disulfonyl-phenyl derivative, OKN-007 suppress free radical formation in experimental cerebral ischemia. OKN-007 failed to show clinical efficacy in stroke, but trials demonstrated its high biosafety in humans including elderly subjects. PBN inhibits the signaling pathways of NF-κB, inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX). In animal models of liver cancer and glioblastoma, OKN-007 seemed more efficient than PBN in suppression of cell proliferation, microvascular density and in induction of apoptosis. OKN-007 also inhibits SULF2 enzyme, which promotes tumor growth via versatile pathways. We assume that nitromemantines may be more beneficial concomitant with chemo-radiotherapy while nitrones alone may act useful in suppressing basal tumor growth and angiogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Memantine/therapeutic use , Nitric Oxide/metabolism , Nitrogen Oxides/therapeutic use , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antineoplastic Agents/chemistry , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Imines/chemistry , Imines/pharmacology , Memantine/analogs & derivatives , Memantine/pharmacology , Models, Biological , Nitrogen Oxides/pharmacology
10.
Nat Commun ; 8(1): 1488, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29133852

ABSTRACT

Transcription factor MEF2C regulates multiple genes linked to autism spectrum disorder (ASD), and human MEF2C haploinsufficiency results in ASD, intellectual disability, and epilepsy. However, molecular mechanisms underlying MEF2C haploinsufficiency syndrome remain poorly understood. Here we report that Mef2c +/-(Mef2c-het) mice exhibit behavioral deficits resembling those of human patients. Gene expression analyses on brains from these mice show changes in genes associated with neurogenesis, synapse formation, and neuronal cell death. Accordingly, Mef2c-het mice exhibit decreased neurogenesis, enhanced neuronal apoptosis, and an increased ratio of excitatory to inhibitory (E/I) neurotransmission. Importantly, neurobehavioral deficits, E/I imbalance, and histological damage are all ameliorated by treatment with NitroSynapsin, a new dual-action compound related to the FDA-approved drug memantine, representing an uncompetitive/fast off-rate antagonist of NMDA-type glutamate receptors. These results suggest that MEF2C haploinsufficiency leads to abnormal brain development, E/I imbalance, and neurobehavioral dysfunction, which may be mitigated by pharmacological intervention.


Subject(s)
Autistic Disorder/genetics , Brain/growth & development , Excitatory Amino Acid Antagonists/therapeutic use , Haploinsufficiency , Memantine/analogs & derivatives , Memantine/therapeutic use , Animals , Autistic Disorder/pathology , Autistic Disorder/physiopathology , Behavior, Animal , Biomarkers/metabolism , Brain/pathology , Brain/physiopathology , Cell Death , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Profiling , Humans , Long-Term Potentiation/genetics , MEF2 Transcription Factors/genetics , Memantine/pharmacology , Mice, Inbred C57BL , Neurogenesis/genetics , Neurons/pathology , Phenotype , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/pathology , Synaptic Transmission/genetics
11.
Eur J Pharm Sci ; 109: 402-411, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28860082

ABSTRACT

Glioblastoma is one of the most aggressive malignant primary brain cancer in adults. To date, surgery, radiotherapy and current pharmacological treatments are not sufficient to manage this pathology that has a high mortality rate (median survival 12-15months). Recently, anticancer multi-targeted compounds have attracted much attention with the aim to obtain new drugs able to hit different biological targets that are involved in the onset and progression of the disease. Here, we report the synthesis of novel memantine-derived drugs (MP1-10) and their potential antitumor activities in human U87MG glioblastoma cell line. MP1-10 were synthetized joining memantine, which is a NMDA antagonist, to different histone deacetylase inhibitors to obtain one molecule with improved therapeutic efficacy. Biological results indicated that MP1 and MP2 possessed more potent anti-proliferative effects on U87MG cells than MP3-10 in a dose-dependent manner. MP1 and MP2 induced significant cell death by apoptosis characterized by apoptotic morphological changes in Hoechst staining. Both drugs also exhibited non-genotoxic and only mild cytotoxic effects in human whole blood cells. However, only MP1, showing good chemico-physical properties (solubility, LogP) and enzymatic stabilities in gastric and intestinal fluids, can be considered a suitable candidate for in vivo pharmacokinetic studies.


Subject(s)
Antineoplastic Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Memantine/analogs & derivatives , Memantine/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Adult , Antineoplastic Agents/chemistry , Blood Cells/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Excitatory Amino Acid Antagonists/chemistry , Gastric Juice/chemistry , Glioblastoma/drug therapy , Histone Deacetylase Inhibitors/chemistry , Humans , Intestinal Mucosa/metabolism , Intestinal Secretions/chemistry , Male , Memantine/chemistry , Solubility , Young Adult
12.
Sci Rep ; 7: 45627, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28358144

ABSTRACT

A new group of compounds, promising for the design of original multitarget therapeutic agents for treating neurodegenerative diseases, based on conjugates of aminoadamantane and carbazole derivatives was synthesized and investigated. Compounds of these series were found to interact with a group of targets that play an important role in the development of this type of diseases. First of all, these compounds selectively inhibit butyrylcholinesterase, block NMDA receptors containing NR2B subunits while maintaining the properties of MK-801 binding site blockers, exert microtubules stabilizing properties, and possess the ability to protect nerve cells from death at the calcium overload conditions. The leading compound C-2h has been shown the most promising effects on all analyzed parameters. Thus, these compounds can be regarded as promising candidates for the design of multi-target disease-modifying drugs for treatment of AD and/or similar neuropathologies.


Subject(s)
Alzheimer Disease/drug therapy , Amantadine/pharmacology , Carbazoles/pharmacology , Memantine/pharmacology , Alzheimer Disease/metabolism , Amantadine/analogs & derivatives , Carbazoles/chemistry , Carboxylesterase/antagonists & inhibitors , Cholinesterase Inhibitors/pharmacology , Drug Design , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Memantine/analogs & derivatives , Microtubules/drug effects , Microtubules/metabolism , Microtubules/ultrastructure , Molecular Docking Simulation , Protein Binding , Receptors, N-Methyl-D-Aspartate/metabolism
13.
Sci Rep ; 5: 14781, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477507

ABSTRACT

Stroke and vascular dementia are leading causes of morbidity and mortality. Neuroprotective therapies have been proposed but none have proven clinically tolerated and effective. While overstimulation of N-methyl-d-aspartate-type glutamate receptors (NMDARs) is thought to contribute to cerebrovascular insults, the importance of NMDARs in physiological function has made this target, at least in the view of many in 'Big Pharma,' 'undruggable' for this indication. Here, we describe novel NitroMemantine drugs, comprising an adamantane moiety that binds in the NMDAR-associated ion channel that is used to target a nitro group to redox-mediated regulatory sites on the receptor. The NitroMemantines are both well tolerated and effective against cerebral infarction in rodent models via a dual allosteric mechanism of open-channel block and NO/redox modulation of the receptor. Targeted S-nitrosylation of NMDARs by NitroMemantine is potentiated by hypoxia and thereby directed at ischemic neurons. Allosteric approaches to tune NMDAR activity may hold therapeutic potential for cerebrovascular disorders.


Subject(s)
Cerebrovascular Disorders/metabolism , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Anura , Apoptosis/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/pathology , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Memantine/analogs & derivatives , Memantine/therapeutic use , Membrane Potentials/drug effects , Nitric Oxide/metabolism , Oxidation-Reduction/drug effects , Rats , Synaptic Transmission/drug effects
14.
Nucl Med Biol ; 42(8): 643-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963911

ABSTRACT

INTRODUCTION: The N-methyl-D-aspartate receptor (NMDAr) is an ionotropic receptor that mediates excitatory transmission. NMDAr overexcitation is thought to be involved in neurological and neuropsychiatric disorders such as Alzheimer disease and schizophrenia. We synthesized [(18)F]-fluoroethylnormemantine ([(18)F]-FNM), a memantine derivative that binds to phencyclidine (PCP) sites within the NMDA channel pore. These sites are primarily accessible when the channel is in the active and open state. METHODS: Radiosynthesis was carried out using the Raytest® SynChrom R&D fluorination module. Affinity of this new compound was determined by competition assay. We ran a kinetic study in rats and computed a time-activity curve based on a volume-of-interest analysis, using CARIMAS® software. We performed an ex vivo autoradiography, exposing frozen rat brain sections to a phosphorscreen. Adjacent sections were used to detect NMDAr by immunohistochemistry with an anti-NR1 antibody. As a control of the specificity of our compound for NMDAr, we used a rat anesthetized with ketamine. Correlation analysis was performed with ImageJ software between signal of autoradiography and immunostaining. RESULTS: Fluorination yield was 10.5% (end of synthesis), with a mean activity of 3145 MBq and a specific activity above 355 GBq/µmol. Affinity assessment allowed us to determine [(19)F]-FNM IC50 at 6.1 10(-6)M. [(18)F]-FMN concentration gradually increased in the brain, stabilizing at 40 minutes post injection. The brain-to-blood ratio was 6, and 0.4% of the injected dose was found in the brain. Combined ex vivo autoradiography and immunohistochemical staining demonstrated colocalization of NMDAr and [(18)F]-FNM (r=0.622, p<0.0001). The highest intensity was found in the cortex and cerebellum, and the lowest in white matter. A low and homogeneous signal corresponding to unspecific binding was observed when PCP sites were blocked with ketamine. CONCLUSIONS: [(18)F]-FNM appears to be a promising tracer for imaging NMDAr activity for undertaking preclinical studies in perspective of clinical detection of neurological or neuropsychological disorders.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Memantine/analogs & derivatives , Memantine/pharmacokinetics , Phencyclidine/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Autoradiography , Fluorine Radioisotopes/pharmacokinetics , Isotope Labeling , Kinetics , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Tissue Distribution
15.
Int J Pharm ; 485(1-2): 183-91, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25747452

ABSTRACT

Solid lipid nanoparticles (SLNs) are considered very attractive drug-delivery systems (DDS) able to enhance the efficacy of some therapeutic agents in several pathologies difficult to treat in a conventional way. Starting from these evidences, this study describes the preparation, physicochemical characterization, release, and in vitro cytotoxicity of stealth SLNs as innovative approach to improve solubility and absorption through the gastrointestinal tract of lipoyl-memantine (LA-MEM), a potential anti-Alzheimer codrug. Physico-chemical properties of LA-MEM loaded SLNs have been intensively investigated. Differential scanning calorimetry (DSC) was used to clarify the state and crystalline structure of the formulation. The results obtained from particles size analysis, polydispersity (PDI), and zeta potential measurements allowed the identification of the optimized formulation, which was characterized by a drug-lipid ratio 1:5, an average intensity diameter of 170nm, a PDI of 0.072, a zeta potential of -33.8mV, and an entrapment efficiency of 88%. Moreover, in vitro stability and release studies in both simulated gastric fluid (SGF) and simulated intestinal fluid (SIF), and preliminary in vitro cytotoxicity studies revealed that LA-MEM loaded SLNs could represent potential candidate for an in vivo investigation as DDS for the brain since it resulted devoid of citotoxicity and able to release the free codrug.


Subject(s)
Drug Carriers , Excitatory Amino Acid Antagonists/chemistry , Lipids/chemistry , Memantine/analogs & derivatives , Memantine/chemistry , Nanoparticles , Thioctic Acid/analogs & derivatives , Animals , Calorimetry, Differential Scanning , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Crystallization , Drug Stability , Excitatory Amino Acid Antagonists/toxicity , Gastric Juice/chemistry , Humans , Intestinal Secretions/chemistry , Kinetics , Memantine/toxicity , Mice , Nanotechnology , Oxidative Stress/drug effects , Particle Size , Solubility , Technology, Pharmaceutical/methods , Thioctic Acid/chemistry , Thioctic Acid/toxicity
16.
Eur J Pharm Sci ; 49(2): 187-98, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23454012

ABSTRACT

The approved treatments for Alzheimer's disease (AD) exploit mainly a symptomatic approach based on the use of cholinesterase inhibitors or N-methyl-D-aspartate (NMDA) receptor antagonists. Natural antioxidant compounds, able to pass through the blood-brain barrier (BBB), have been extensively studied as useful neuroprotective agents. A novel approach towards excitotoxicity protection and oxidative stress associated with excess ß amyloid (Aß) preservation in AD is represented by selective glutamatergic antagonists that possess as well antioxidant capabilities. In the present work, GSH (1) or (R)-α-lipoic acid (LA) (2) have been covalently linked with the NMDA receptor antagonists memantine (MEM). The new conjugates, proposed as potential antialzheimer drugs, should act both as glutamate receptor antagonists and radical scavenging agents. The physico-chemical properties and "in vitro" membrane permeability, the enzymatic and chemical stability, the demonstrated "in vitro" antioxidant activity associated to the capacity to inhibit Aß(1-42) aggregation makes at least compound 2 a promising candidate for treatment of AD patients.


Subject(s)
Antioxidants/chemistry , Excitatory Amino Acid Antagonists/chemistry , Glutathione/chemistry , Memantine/analogs & derivatives , Memantine/chemistry , Prodrugs/chemistry , Thioctic Acid/analogs & derivatives , Thioctic Acid/chemistry , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Animals , Antioxidants/pharmacology , Cell Line , Excitatory Amino Acid Antagonists/pharmacology , Glutathione/pharmacology , Hippocampus/cytology , Humans , Male , Memantine/pharmacology , Membranes, Artificial , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Norepinephrine/metabolism , Peptide Fragments/chemistry , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptosomes/drug effects , Synaptosomes/metabolism , Thioctic Acid/pharmacology
17.
Nat Rev Drug Discov ; 5(2): 160-70, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16424917

ABSTRACT

Neuroprotective drugs tested in clinical trials, particularly those that block N-methyl-D-aspartate-sensitive glutamate receptors (NMDARs), have failed miserably in large part because of intolerable side effects. However, one such drug, memantine, was recently approved by the European Union and the US FDA for the treatment of dementia following our group's discovery of its clinically tolerated mechanism of action. Here, we review the molecular basis for memantine efficacy in neurological diseases that are mediated, at least in part, by overactivation of NMDARs, producing excessive Ca(2+) influx through the receptor's associated ion channel and consequent free-radical formation.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Humans , Memantine/analogs & derivatives , Neurotoxins/pharmacology
18.
Biomed Chromatogr ; 20(5): 423-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16161183

ABSTRACT

Simultaneous HPLC assay of 1-adamantanamine hydrochloride (amantadine) and its four related compounds [2-adamantanamine hydrochloride (2-ADA), 1-adamantanmethylamine (ADAMA), 1-(1-adamantyl)ethylamine hydrochloride (rimantadine) and 3,5-dimethyl-1-adamantanamine hydrochloride (memantine)] in phosphate-buffered saline (pH 7.4) after pre-column derivatization with 4-fluoro-7-nitro-2,1,3-benzoxadiazole (NBD-F) was developed. Phosphate-buffered saline samples were mixed with borate buffer and NBD-F solution in acetonitrile at 60 degrees C for 5 min and injected into HPLC. Five derivatives were well separated from each other. The lower limits of detection of amantadine, 2-ADA, ADAMA, rimantadine and memantine were 0.008, 0.001, 0.0008, 0.0015 and 0.01 microg/mL, respectively. The coefficients of variation for intra- and inter-day assay were less than 6.4 and 8.2%, respectively. The method presented was applied to a binding study of these compounds to human alpha(1)-acid glycoprotein. While affinity constants and capacities for ADAMA, rimantadine and memantine were calculated by means of Scatchard plots, those for the others were not determined. ADAMA, rimantadine and memantine were bound with different affinities and capacities. These results indicate that NBD-F is a good candidate as a fluorescent reagent to simultaneously determine amantadine and its four related compounds by HPLC after pre-column derivatization. Our method can be applied to binding studies for protein.


Subject(s)
Amantadine/isolation & purification , Chromatography, High Pressure Liquid/methods , 4-Chloro-7-nitrobenzofurazan/chemistry , Amantadine/analogs & derivatives , Amantadine/chemistry , Binding, Competitive , Calibration , Fluorescent Dyes/chemistry , Humans , Memantine/analogs & derivatives , Memantine/chemistry , Memantine/isolation & purification , Phosphates/chemistry , Reproducibility of Results , Rimantadine/analogs & derivatives , Rimantadine/chemistry , Rimantadine/isolation & purification , Sodium Chloride/chemistry
20.
Nat Neurosci ; 8(4): 527-33, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15768036

ABSTRACT

Formation of senile plaques composed of amyloid beta peptide, a pathological hallmark of Alzheimer disease, in human brains precedes disease onset by many years. Noninvasive detection of such plaques could be critical in presymptomatic diagnosis and could contribute to early preventive treatment strategies. Using amyloid precursor protein (APP) transgenic mice as a model of amyloid beta amyloidosis, we demonstrate here that an intravenously administered (19)F-containing amyloidophilic compound labels brain plaques and allows them to be visualized in living mice by magnetic resonance imaging (MRI) using (19)F and (1)H. Our findings provide a new direction for specific noninvasive amyloid imaging without the danger of exposure to radiation. This approach could be used in longitudinal studies in mouse models of Alzheimer disease to search for biomarkers associated with amyloid beta pathology as well as to track disease course after treatment with candidate medications.


Subject(s)
Amyloid beta-Peptides/analysis , Amyloidosis/diagnosis , Magnetic Resonance Imaging , Memantine/analogs & derivatives , Tritium , Age Factors , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/metabolism , Animals , Brain/diagnostic imaging , Brain/pathology , Brain Chemistry/physiology , Cell Count/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Humans , Imaging, Three-Dimensional/methods , Immunohistochemistry/methods , Magnetic Resonance Imaging/methods , Memantine/pharmacokinetics , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/metabolism , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/pathology , Radionuclide Imaging , Staining and Labeling/methods , Time Factors , Tritium/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...