Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Amino Acids ; 53(9): 1351-1359, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34283312

ABSTRACT

The cyanobacterial non-protein amino acid α-amino-ß-methylaminopropionic acid, more commonly known as BMAA, was first discovered in the seeds of the ancient gymnosperm Cycad circinalis (now Cycas micronesica Hill). BMAA was linked to the high incidence of neurological disorders on the island of Guam first reported in the 1950s. BMAA still attracts interest as a possible causative factor in amyotrophic lateral sclerosis (ALS) following the identification of ALS disease clusters associated with living in proximity to lakes with regular cyanobacterial blooms. Since its discovery, BMAA toxicity has been the subject of many in vivo and in vitro studies. A number of mechanisms of toxicity have been proposed including an agonist effect at glutamate receptors, competition with cysteine for transport system xc_ and other mechanisms capable of generating cellular oxidative stress. In addition, a wide range of studies have reported effects related to disturbances in proteostasis including endoplasmic reticulum stress and activation of the unfolded protein response. In the present studies we examine the effects of BMAA on the ubiquitin-proteasome system (UPS) and on chaperone-mediated autophagy (CMA) by measuring levels of ubiquitinated proteins and lamp2a protein levels in a differentiated neuronal cell line exposed to BMAA. The BMAA induced increases in oxidised proteins and the increase in CMA activity reported could be prevented by co-administration of L-serine but not by the two antioxidants examined. These data provide further evidence of a protective role for L-serine against the deleterious effects of BMAA.


Subject(s)
Amino Acids, Diamino/adverse effects , Chaperone-Mediated Autophagy , Cyanobacteria Toxins/adverse effects , Lysosomal-Associated Membrane Protein 2/metabolism , Neuroblastoma/drug therapy , Protein Aggregates/drug effects , Serine/pharmacology , Ubiquitin/metabolism , Antioxidants/pharmacology , Cell Differentiation , Excitatory Amino Acid Agonists/adverse effects , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oxidative Stress , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Tumor Cells, Cultured
2.
Pharmacol Biochem Behav ; 203: 173154, 2021 04.
Article in English | MEDLINE | ID: mdl-33609599

ABSTRACT

Interest in developing NMDA receptor antagonists with reduced side-effects for neurological and psychiatric disorders has been re-energized by the recent introduction of esketamine into clinical practice for treatment-resistant depression. Structural analogs of dextromethorphan bind with low affinity to the NMDA receptor ion channel, have functional effects in vivo, and generally display a lower propensity for side-effects than that of ketamine and other higher affinity antagonists. As such, the aim of the present study was to determine whether a series of N-substituted-3-alkoxy-substituted dextromethorphan analogs produce their anticonvulsant effects through NMDA receptor blockade. Compounds were studied against NMDA-induced seizures in rats. Compounds were administered intracerebroventricularly in order to mitigate confounds of drug metabolism that arise from systemic administration. Comparison of the anticonvulsant potencies to their affinities for NMDA, σ1, and σ2 binding sites were made in order to evaluate the contribution of these receptors to anticonvulsant efficacy. The potencies to block convulsions were positively associated with their affinities to bind to the NMDA receptor ion channel ([3H]-TCP binding) (r = 0.71, p < 0.05) but not to σ1 receptors ([3H]-SKF 10047 binding) (r = -0.31, p = 0.46) or to σ2 receptors ([3H]-DTG binding) (p = -0.38, p = 0.36). This is the first report demonstrating that these dextromethorphan analogs are functional NMDA receptor antagonists in vivo. Given their potential therapeutic utility and favorable side-effect profiles, such low affinity NMDA receptor antagonists could be considered for further development in neurological (e.g., anticonvulsant) and psychiatric (e.g., antidepressant) disorders.


Subject(s)
Anticonvulsants/administration & dosage , Dextromethorphan/analogs & derivatives , Dextromethorphan/administration & dosage , Dextrorphan/administration & dosage , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/administration & dosage , N-Methylaspartate/adverse effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/chemically induced , Seizures/drug therapy , Alcohols/chemistry , Animals , Anticonvulsants/metabolism , Binding Sites , Dextromethorphan/metabolism , Dextrorphan/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/metabolism , Infusions, Intraventricular , Ligands , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Treatment Outcome , Sigma-1 Receptor
3.
Neuropharmacology ; 148: 87-95, 2019 04.
Article in English | MEDLINE | ID: mdl-30597159

ABSTRACT

Experimental evidence indicates that anesthetic doses of the non-competitive NMDA receptor antagonist ketamine impair memory abilities in rodents. The mechanism by which anesthetic ketamine produces its adverse behavioural effects is not yet clarified. In this context, it has been proposed that the effects of anesthetic ketamine on memory might be attributed to its agonistic properties on the GABA type A receptor. The present study was designed to address this issue. Thus, we investigated the ability of the benzodiazepine receptor antagonist flumazenil (1, 3, 6 mg/kg, i.p.) and the GABAA receptor antagonist bicuculline (0.5, 1.5, 3 mg/kg, i.p.) to counteract recognition memory deficits produced by anesthetic ketamine (100 mg/kg, i.p.) in rats. For this purpose, the novel object recognition task, a behavioural paradigm assessing recognition memory abilities in rodents was used. Compounds were coadministered 24 h before testing or retention. Pre (24 h before testing) or post-training (24 h before retention) administration of flumazenil (6 mg/kg, i.p.) counteracted anesthetic ketamine-induced performance deficits in the novel object recognition memory task. Conversely, bicuculline failed to attenuate the recognition memory deficits caused by anesthetic ketamine. Our findings propose a functional interaction between anesthetic ketamine and the GABAA receptor allosteric modulator flumazenil on recognition memory.


Subject(s)
Bicuculline/pharmacology , Flumazenil/pharmacology , Ketamine/adverse effects , Memory Disorders/prevention & control , Recognition, Psychology/drug effects , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Ketamine/antagonists & inhibitors , Male , Memory Disorders/chemically induced , Rats
4.
Chemosphere ; 214: 623-632, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30290362

ABSTRACT

The non-proteinogenic amino acid ß-N-methylamino-l-alanine (BMAA) is associated with the development of neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis/parkinsonism-dementia complex (ALS-PDC) and amyotrophic lateral sclerosis. BMAA is known to induce neurotoxic effects leading to neurodegeneration via multiple mechanisms including misfolded protein accumulation, glutamate induced excitotoxicity, calcium dyshomeostasis, endoplasmic reticulum stress and oxidative stress. In the present study, for the first time, genotoxic activity of BMAA (2.5, 5, 10 and 20 µg/mL) was studied in human peripheral blood cells (HPBCs) using the comet and cytokinesis-block micronucleus cytome assays. In addition, the influence of BMAA on the oxidative stress was assessed. At non-cytotoxic concentrations BMAA did not induce formation of DNA strand breaks in HPBCs after 4 and 24 h exposure; however, it significantly increased the number of micronuclei after 24 and 48 h at 20 µg/mL and nucleoplasmic bridges after 48 h at 20 µg/mL. The frequency of nuclear buds was slightly though non-significantly increased after 48 h. Altogether, this indicates that in HPBCs BMAA is clastogenic and induces complex genomic alterations including structural chromosomal rearrangements and gene amplification. No influence on oxidative stress markers was noticed. These findings provide new evidence that environmental neurotoxin BMAA, in addition to targeting common pathways involved in neurodegeneration, can also induce genomic instability in non-target HPBCs suggesting that it might be involved in cancer development. Therefore, these data are important in advancing our current knowledge and opening new questions in the understanding of the mechanisms of BMAA toxicity, particularly in the context of genotoxicity.


Subject(s)
Amino Acids, Diamino/adverse effects , Biomarkers/metabolism , Blood Cells/pathology , Neurotoxins/adverse effects , Oxidative Stress/drug effects , Adult , Blood Cells/drug effects , Blood Cells/metabolism , Cyanobacteria Toxins , DNA Damage , Endoplasmic Reticulum Stress/drug effects , Excitatory Amino Acid Agonists/adverse effects , Female , Humans
5.
Sci Rep ; 7(1): 15146, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29123180

ABSTRACT

Many neurological disorders of gluten-related diseases (GRD), not directly referable to the gastrointestinal tract, have been reported in association with celiac disease (CD), including ataxia, neuropathy and epilepsy. In particular, people with epilepsy diagnosed with CD seems to be characterized by intractable seizure. In these patients, gluten restriction diet has resulted in a reduction of both seizure frequency and antiepileptic medication. Many hypotheses have been suggested, however, molecular mechanisms that associates GRD and epileptogenesis are yet unknown. In this study, we examined the effects of the toxic gliadin peptide 31-43 in in vivo and in vitro models of kainate-induced-epilepsy. We observed that p31-43 exacerbates kainate neurotoxicity in epilepsy models, through the involvement of the enzymatic activity of transglutaminases. Moreover, electrophysiological recordings in CA3 pyramidal neurons of organotypic hippocampal slices show that p31-43 increases the inward current induced by kainate, the average sEPSC amplitude and the total number of evoked action potentials when applicated alone, thus suggesting that p31-43 is able to influence CA3-CA1 neurotransmission and can potentiate postsynaptic kainate receptors. Our results suggest a possible mechanism underlying the relationship between GRD and epilepsy through a potentiation of kainate-induced neurotoxicity and links the toxic effects of gluten to epilepsy.


Subject(s)
Celiac Disease/complications , Epilepsy/chemically induced , Epilepsy/pathology , Excitatory Amino Acid Agonists/adverse effects , Gliadin/metabolism , Kainic Acid/adverse effects , Peptide Fragments/metabolism , Action Potentials , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Electroencephalography , Excitatory Amino Acid Agonists/metabolism , Humans , Kainic Acid/metabolism , Transglutaminases/metabolism
6.
Epilepsia ; 57(6): 967-76, 2016 06.
Article in English | MEDLINE | ID: mdl-27173148

ABSTRACT

OBJECTIVE: Systemic administration of kainic acid (KA) is a widely used procedure utilized to develop a model of temporal lobe epilepsy (TLE). Despite its ability to induce status epilepticus (SE) in vivo, KA applied to in vitro preparations induces only interictal-like activity and/or isolated ictal discharges. The possibility that extravasation of the serum protein albumin from the vascular compartment enhances KA-induced brain excitability is investigated here. METHODS: Epileptiform activity was induced by arterial perfusion of 6 µm KA in the in vitro isolated guinea pig brain preparation. Simultaneous field potential recordings were carried out bilaterally from limbic (CA1, dentate gyrus [DG], and entorhinal cortex) and extralimbic regions (piriform cortex and neocortex). Blood-brain barrier (BBB) breakdown associated with KA-induced epileptiform activity was assessed by parenchymal leakage of intravascular fluorescein-isothiocyanate albumin. Seizure-induced brain inflammation was evaluated by western blot analysis of interleukin (IL)-1ß expression in brain tissue. RESULTS: KA infusion caused synchronized activity at 15-30 Hz in limbic (but not extralimbic) cortical areas, associated with a brief, single seizure-like event. A second bolus of KA, 60 min after the induction of the first ictal event, did not further enhance excitability. Perfusion of serum albumin between the two administrations of KA enhanced epileptiform discharges and allowed a recurrent ictal event during the second KA infusion. SIGNIFICANCE: Our data show that arterial KA administration selectively alters the synchronization of limbic networks. However, KA is not sufficient to generate recurrent seizures unless serum albumin is co-perfused during KA administration. These findings suggest a role of serum albumin in facilitating acute seizure generation.


Subject(s)
Albumins/metabolism , Blood-Brain Barrier/drug effects , Excitatory Amino Acid Agonists/adverse effects , Kainic Acid/adverse effects , Limbic System/physiopathology , Seizures/chemically induced , Animals , Capillary Permeability/drug effects , Disease Models, Animal , Electroencephalography , Female , Glial Fibrillary Acidic Protein/metabolism , Guinea Pigs , Interleukin-1beta/metabolism , Limbic System/drug effects , Microscopy, Confocal , Phosphopyruvate Hydratase/metabolism , Serum Albumin/pharmacology , Spectrum Analysis , Subcellular Fractions/metabolism
7.
Mol Autism ; 7: 2, 2016.
Article in English | MEDLINE | ID: mdl-26770664

ABSTRACT

BACKGROUND: Researchers have demonstrated that d-cycloserine (DCS) can enhance the effects of behavioral interventions in adults with anxiety and enhances prosocial behavior in animal models of autism spectrum disorders (ASD). This study extended upon this background by combining DCS with behavioral social skills therapy in youth with ASD to assess its impact on the core social deficits of ASD. We hypothesized that DCS used in combination with social skills training would enhance the acquisition of social skills in children with ASD. METHODS: A 10-week, double-blind, placebo-controlled trial of DCS (50 mg) given 30 min prior to weekly group social skills training was conducted at two sites. Children with ASD were randomized to receive 10 weeks (10 doses) of DCS or placebo in a 1:1 ratio. RESULTS: No statistically significant difference attributable to drug treatment was observed in the change scores for the primary outcome measure, the Social Responsiveness Scale (SRS), total score (p = 0.45), or on secondary outcome measures. CONCLUSIONS: The results of this trial demonstrated no drug-related short-term improvement on the primary outcome measure, or any of the secondary outcome measures. However, an overall significant improvement in SRS total raw score was observed from baseline to end of treatment for the entire group of children with ASD. This suggests a need to further study the efficacy of the social skills training protocol. Limitations to the current study and areas for future research are discussed. TRIAL REGISTRATION: ClinicalTrials.govNCT01086475.


Subject(s)
Autism Spectrum Disorder/drug therapy , Behavior Therapy , Cycloserine/therapeutic use , Excitatory Amino Acid Agonists/therapeutic use , Social Skills , Autism Spectrum Disorder/psychology , Autism Spectrum Disorder/therapy , Child , Child, Preschool , Cycloserine/adverse effects , Double-Blind Method , Excitatory Amino Acid Agonists/adverse effects , Female , Humans , Interpersonal Relations , Learning/drug effects , Male , Parents/psychology , Severity of Illness Index , Treatment Failure
8.
Article in English | MEDLINE | ID: mdl-26364274

ABSTRACT

D-Cycloserine, known from tuberculosis therapy, has been widely introduced to neuropsychiatric studies, since its central active mechanism as a partial NMDA-agonist has been found. In this review, we evaluate its therapeutic potential in neuropsychological disorders and discuss its pitfalls in terms of dosing and application frequency as well as its safety in low-dose therapy. Therefore, we identified 91 clinical trials by performing a Medline search. We demonstrate in part preliminary but increasing evidence that D-cycloserine may be effective in various psychiatric diseases, including schizophrenia, anxiety disorders, addiction, eating disorders, major depression, and autism as well as in neurological diseases, including dementia, Alzheimer's disease, and spinocerebellar degeneration. D-Cycloserine in low-dose therapy is safe, but there is still a need for new drugs with higher specificity to the different N-methyl-D-aspartate-receptor subunits.


Subject(s)
Cycloserine/therapeutic use , Excitatory Amino Acid Agonists/therapeutic use , Mental Disorders/drug therapy , Nervous System Diseases/drug therapy , Psychotropic Drugs/therapeutic use , Receptors, N-Methyl-D-Aspartate/agonists , Animals , Cycloserine/adverse effects , Cycloserine/pharmacology , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , Humans , Mental Disorders/metabolism , Nervous System Diseases/metabolism , Psychotropic Drugs/adverse effects , Psychotropic Drugs/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
9.
PLoS One ; 10(12): e0144017, 2015.
Article in English | MEDLINE | ID: mdl-26658273

ABSTRACT

G-protein-coupled receptor (GPCR) agonists are known to induce both cellular adaptations resulting in tolerance to therapeutic effects and withdrawal symptoms upon treatment discontinuation. Glutamate neurotransmission is an integral part of sleep-wake mechanisms, which processes have translational relevance for central activity and target engagement. Here, we investigated the efficacy and tolerance potential of the metabotropic glutamate receptors (mGluR2/3) agonist LY354740 versus mGluR2 positive allosteric modulator (PAM) JNJ-42153605 on sleep-wake organisation in rats. In vitro, the selectivity and potency of JNJ-42153605 were characterized. In vivo, effects on sleep measures were investigated in rats after once daily oral repeated treatment for 7 days, withdrawal and consecutive re-administration of LY354740 (1-10 mg/kg) and JNJ-42153605 (3-30 mg/kg). JNJ-42153605 showed high affinity, potency and selectivity at mGluR2. Binding site analyses and knowledge-based docking confirmed the specificity of JNJ-42153605 at the mGluR2 allosteric binding site. Acute LY354740 and JNJ-42153605 dose-dependently decreased rapid eye movement (REM) sleep time and prolonged its onset latency. Sub chronic effects of LY354740 on REM sleep measures disappeared from day 3 onwards, whereas those of JNJ-42153605 were maintained after repeated exposure. LY354740 attenuated REM sleep homeostatic recovery, while this was preserved after JNJ-42153605 administration. JNJ-42153605 enhanced sleep continuity and efficiency, suggesting its potential as an add-on medication for impaired sleep quality during early stages of treatment. Abrupt cessation of JNJ-42153605 did not induce withdrawal phenomena and sleep disturbances, while the initial drug effect was fully reinstated after re-administration. Collectively, long-term treatment with JNJ-42153605 did not induce tolerance phenomena to its primary functional effects on sleep measures, nor adverse effects at withdrawal, while it promoted homeostatic recovery sleep. From the translational perspective, the present rodent findings suggest that mGluR2 positive allosteric modulation has therapeutic potential based on its superior long term efficacy over agonists in psychiatric disorders, particularly of those commonly occurring with REM sleep overdrive.


Subject(s)
Bridged Bicyclo Compounds/adverse effects , Drug Tolerance , Excitatory Amino Acid Agonists/adverse effects , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/agonists , Sleep, REM/drug effects , Triazines/pharmacology , Allosteric Regulation , Allosteric Site/drug effects , Animals , Binding, Competitive , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetulus , Drug Administration Schedule , Gene Expression , Humans , Ligands , Male , Molecular Docking Simulation , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sleep, REM/physiology , Structural Homology, Protein , Wakefulness/physiology
10.
Int J Clin Pharmacol Ther ; 53(6): 462-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25881753

ABSTRACT

OBJECTIVE: This thorough QT/QTc (TQT) study assessed the effects of a supratherapeutic dose of pomaglumetad methionil, a potential treatment for schizophrenia, compared to placebo on the QT interval in subjects with schizophrenia. METHODS: This double-blind, 3-period crossover study enrolled 86 subjects aged 22 - 63 years, who met Diagnostic and Statistical Manual, Fourth Edition, Test Revision (DSM-IV-TR) criteria for schizophrenia; 78 subjects completed the study. Subjects were randomly assigned to sequences of 3 treatment periods of single oral doses of pomaglumetad methionil 400 mg, moxifloxacin 400 mg, and placebo. Quadruplicate electrocardiograms (ECGs) were extracted from 2 hours predose to 12 hours postdose and were overread by a blinded central reader. Time-matched pharmacokinetic (PK) parameters were assessed. RESULTS: At all-time points, the upper bound of the 90% 2-sided confidence interval (CI) for the least squares (LS) mean difference in changes from baseline in Fridericia's corrected QT interval (ΔQTcF) between pomaglumetad methionil and placebo was < 10 milliseconds (msec). Sufficient assay sensitivity was not achieved, likely due to food effect; although the maximum observed drug concentration (Cmax) with moxifloxacin (1,410 ng/mL) was lower than expected, the slope of the regression line of moxifloxacin plasma concentrations versus placebo-subtracted ΔQTcF was similar to that reported in the literature. CONCLUSIONS: A single supratherapeutic dose of 400 mg pomaglumetad methionil did not prolong QTcF to a clinically significant degree and, importantly, did not result in any absolute QTcF > 450 msec or increase in QTcF from predose > 30 msec.


Subject(s)
Amino Acids/administration & dosage , Excitatory Amino Acid Agonists/administration & dosage , Heart Rate/drug effects , Prodrugs/administration & dosage , Schizophrenia/drug therapy , Administration, Oral , Adult , Amino Acids/adverse effects , Amino Acids/blood , Amino Acids/pharmacokinetics , Cross-Over Studies , Double-Blind Method , Drug Monitoring , Electrocardiography , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/blood , Excitatory Amino Acid Agonists/pharmacokinetics , Female , Humans , Least-Squares Analysis , Male , Middle Aged , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Risk Assessment , Schizophrenia/blood , Schizophrenia/diagnosis , Schizophrenic Psychology , Treatment Outcome , United States , Young Adult
11.
J Psychiatr Pract ; 21(2): 140-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25782764

ABSTRACT

BACKGROUND: Approximately 45% of patients with major depressive disorder (MDD) do not remit when treated with biogenic amine antidepressants. Consequently, there is a significant need for antidepressant agents with different mechanisms of action. Early proof of concept (POC) studies with such novel agents play a significant role in helping drug developers identify agents and mechanisms of action that merit more intensive research. Studies have demonstrated that high affinity N-methyl-Daspartate (NMDA) receptor blockers (eg, ketamine) can produce rapid antidepressant effects in patients who have not responded to currently available agents, but treatment with these agents is accompanied by psychotomimetic effects that make their use problematic. This column describes a POC study involving GLYX-13, an N-methyl-D-aspartate receptor glycine site functional partial agonist. METHOD: In this double-blind, randomized, placebo-controlled study, a single intravenous (IV) dose of GLYX-13 (1, 5, 10, or 30 mg/kg) or placebo was administered to 116 subjects with MDD who had not benefitted from a trial of at least one biogenic amine antidepressant during the current episode. The primary outcome measure was score on the Hamilton Depression Rating Scale-17 (Ham-D17), which was used to rate overall depressive symptoms at baseline and at 24 hours and days 3, 7, 14, and, in some arms, days 21 and 28 after administration. RESULTS: GLYX-13, 5 or 10 mg/kg IV, reduced depressive symptoms as assessed by the Ham-D17 at days 1 through 7. Onset of action as assessed using the Bech-6 occurred within 2 hours. GLYX-13 did not elicit psychotomimetic or other significant side effects. CONCLUSION: In this early POC study, GLYX-13 reduced depressive symptoms within 2 hours and this effect was maintained for 7 days on average in subjects with MDD who had not responded to another antidepressant agent during the current depressive episode. The findings of this study support the hypothesis that modulation of the NMDA receptor is a valid target for the development of antidepressant drugs and the need for additional studies to further evaluate the effects of GLYX-13. POC studies such as the one described here play a pivotal role in allowing drug researchers to decide whether to move forward with larger and more expensive studies, and they enable them to focus available resources on those molecules that appear to have the most therapeutic promise. Based on the POC study described here, a multiple dose study has been completed which showed sustained therapeutic benefit with repeated dosing of GLYX-13 for more than 6 weeks. Phase 3 studies are now being planned.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder, Major/drug therapy , Excitatory Amino Acid Agonists/pharmacology , Oligopeptides/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Adult , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Double-Blind Method , Drug Discovery , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/adverse effects , Female , Glycine/metabolism , Humans , Male , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Treatment Outcome , Young Adult
12.
Steroids ; 98: 166-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25578735

ABSTRACT

Imbalance of excitation and inhibition in neurons is implicated in the pathogenesis of epilepsy. Voltage-gated sodium channels, which play a vital role in regulating neuronal excitability, are one of the major targets for developing anti-epileptic drugs. Here we provide evidence that cholestane-3ß,5α,6ß-triol (triol), a major metabolic oxysterol of cholesterol, is an effective state-dependent negative sodium channels modulator. Triol reduced Na(+) current density in a concentration-dependent manner. 10 µM triol shifted steady-state/fast/slow inactivation curves of sodium channels toward the hyperpolarizing direction. Additionally, triol reduced voltage-gated sodium currents in a voltage- and frequency-dependent manner. In a kainic acid-induced seizures mouse model, triol (25 mg/kg) significantly increased the latency of seizure onset and attenuated seizure severity. Our findings provide novel insights for understanding the modulatory role of a small molecular oxysterol on voltage-gated sodium channels and suggest triol may represent a novel and promising candidate for epilepsy intervention.


Subject(s)
Anticonvulsants/pharmacology , Cholestanols/pharmacology , Epilepsy/drug therapy , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism , Animals , Anticonvulsants/chemistry , Cholestanols/chemistry , Cholesterol/chemistry , Cholesterol/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy/chemically induced , Epilepsy/metabolism , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/adverse effects , Kainic Acid/pharmacology , Mice , Voltage-Gated Sodium Channel Blockers/chemistry
13.
Curr Opin Pharmacol ; 20: 1-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25462286

ABSTRACT

The metabotropic glutamate receptor family includes many potential therapeutic targets for a wide range of neurological disorders however to date no approved drugs have progressed to market. For some receptor subtypes it has been difficult to separate therapeutic benefit from undesirable side effects. For others finding suitable drug like molecules has been challenging. Chemotypes identified from screening have been limited and difficult to optimise away from undesirable groups. Frequently within related series, compounds have switched from agonist to antagonists. Recently the structures of the transmembrane domain of mGlu1 and mGlu5 have been solved revealing the binding site of allosteric modulators which provides an understanding of the difficulties to date and an opportunity for future structure based approaches to drug design.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Metabotropic Glutamate/chemistry , Allosteric Regulation/drug effects , Binding Sites , Drug Design , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/adverse effects , Humans , Molecular Targeted Therapy , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors
14.
Cytotherapy ; 16(12): 1760-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25442789

ABSTRACT

BACKGROUND AIMS: Hippocampal neurodegeneration is one of the hallmarks in neurological and neurodegenerative diseases such as temporal lobe epilepsy and Alzheimer disease. Human embryonic kidney (HEK) cells are a mixed population of cells, including neurons, and their conditioned medium is enriched with erythropoietin (EPO). Because EPO is a known neuroprotectant, we hypothesized that infusion of HEK cells or HEK-conditioned medium (HEK-CM) may provide neuroprotection against kainic acid (KA)-induced hippocampal damage in mice. METHODS: Adult CF1 mice were treated with KA to induce hippocampal damage. On 3rd and 5th days after KA treatment, HEK cells or HEK-CM was infused intravenously through the tail vein. On the 7th and 8th days after KA treatment, all groups of mice were subjected to cognitive and depression assessment by use of a novel object recognition test and a forced swim test, respectively. Subsequent to this assessment, mice were killed and the brain samples were used to assess the histopathology and messenger RNA expression for EPO and B-cell lymphoma-2 (Bcl-2). RESULTS: We found that infusion of HEK cells/HEK-CM improves cognitive function and alleviates symptoms of depression. Histological assessment demonstrates complete neuroprotection against KA-mediated excitotoxicity, and the hippocampal cytoarchitecture of HEK cells/HEK-CM treated mice was comparable to normal control mice. HEK cells/HEK-CM treatment could provide neuroprotection by upregulating the endogenous EPO and Bcl-2 in KA-treated mice. CONCLUSIONS: Our present data demonstrate for the first time that infusion of HEK cells/HEK-CM can prevent excitotoxic hippocampal damage and alleviate consequent behavioral abnormalities.


Subject(s)
Brain Injuries , Culture Media, Conditioned , Excitatory Amino Acid Agonists/adverse effects , Hippocampus/injuries , Kainic Acid/adverse effects , Animals , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/metabolism , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Excitatory Amino Acid Agonists/pharmacology , HEK293 Cells , Hippocampus/metabolism , Humans , Kainic Acid/pharmacology , Male , Mice , Time Factors
15.
Expert Opin Investig Drugs ; 23(9): 1181-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24818801

ABSTRACT

INTRODUCTION: Mood disorders, including depression, are becoming increasingly prevalent in the developed world. Furthermore, treatment of depression therapeutics, mainly influencing the serotonergic and adrenergic systems, is considered insufficient. The original NMDA-glutamate hypothesis mechanism of antidepressant action was first proposed ∼ 20 years ago. Since then, a number of preclinical and clinical studies have examined its rationale. AREAS COVERED: This review highlights the recent clinical evidence for the use of functional NMDA receptor antagonists as antidepressants. Furthermore, the authors present the mechanism(s) of antidepressant action derived mostly from preclinical paradigms. EXPERT OPINION: Currently, clinical studies mostly use ketamine (a noncompetitive high-potency NMDA antagonist) as an agent for rapid relief of depressive symptoms. However, due to the ketamine-induced psychotomimetic effects, new NMDA receptor antagonists (modulators) are continuously being introduced for rapid antidepressant action, especially for use in treatment-resistant patients. Recent clinical reports for the use of CP-101,606, MK-0657 (selective GluN2B subunit NMDA receptor antagonists), GLYX-13 and d-cycloserine (glycine site partial agonists) are optimistic but await further support.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Excitatory Amino Acid Agonists/therapeutic use , Animals , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacology , Depressive Disorder, Major/physiopathology , Drug Design , Drug Resistance , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , Humans , Ketamine/administration & dosage , Ketamine/pharmacology , Ketamine/therapeutic use , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
16.
PLoS One ; 9(5): e95318, 2014.
Article in English | MEDLINE | ID: mdl-24798187

ABSTRACT

BACKGROUND: Dietary long-chain n-3 polyunsaturated fatty acid (PUFA) supplementation may be beneficial for chronic brain illnesses, but the issue is not agreed on. We examined effects of dietary n-3 PUFA deprivation or supplementation, compared with an n-3 PUFA adequate diet (containing alpha-linolenic acid [18:3 n-3] but not docosahexaenoic acid [DHA, 22:6n-3]), on brain markers of lipid metabolism and excitotoxicity, in rats treated chronically with NMDA or saline. METHODS: Male rats after weaning were maintained on one of three diets for 15 weeks. After 12 weeks, each diet group was injected i.p. daily with saline (1 ml/kg) or a subconvulsive dose of NMDA (25 mg/kg) for 3 additional weeks. Then, brain fatty acid concentrations and various markers of excitotoxicity and fatty acid metabolism were measured. RESULTS: Compared to the diet-adequate group, brain DHA concentration was reduced, while n-6 docosapentaenoic acid (DPA, 22:5n-6) concentration was increased in the n-3 deficient group; arachidonic acid (AA, 20:4n-6) concentration was unchanged. These concentrations were unaffected by fish oil supplementation. Chronic NMDA increased brain cPLA2 activity in each of the three groups, but n-3 PUFA deprivation or fish oil did not change cPLA2 activity or protein compared with the adequate group. sPLA2 expression was unchanged in the three conditions, whereas iPLA2 expression was reduced by deprivation but not changed by supplementation. BDNF protein was reduced by NMDA in N-3 PUFA deficient rats, but protein levels of IL-1ß, NGF, and GFAP did not differ between groups. CONCLUSIONS: N-3 PUFA deprivation significantly worsened several pathological NMDA-induced changes produced in diet adequate rats, whereas n-3 PUFA supplementation did not affect NMDA induced changes. Supplementation may not be critical for this measured neuropathology once the diet has an adequate n-3 PUFA content.


Subject(s)
Brain Diseases/metabolism , Dietary Fats/adverse effects , Excitatory Amino Acid Agonists/adverse effects , Fatty Acids, Omega-3/adverse effects , Lipid Metabolism/drug effects , N-Methylaspartate/adverse effects , Animals , Brain Chemistry/drug effects , Brain Diseases/chemically induced , Brain Diseases/pathology , Chronic Disease , Dietary Fats/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Fatty Acids, Omega-3/pharmacology , Group IV Phospholipases A2/metabolism , Interleukin-1beta/metabolism , Male , N-Methylaspartate/pharmacology , Nerve Growth Factor/metabolism , Rats
17.
J Pharm Pharmacol ; 66(9): 1294-302, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24707860

ABSTRACT

OBJECTIVES: Aloysia gratissima aqueous extract (AE) was investigated as a putative protective agent against quinolinic acid (QA)-induced seizures in mice and hippocampal cell damage. Additionally, AE and ferulic acid (FA), the major compound of AE, were tested against neurotoxicity evoked by glutamate or its N-methyl-D-aspartate receptor (NMDAR) agonist, QA on hippocampal slices, in vitro. METHODS: Mice were treated with AE before QA infusion (36.8 nmol/site) and seizures were analysed. Cellular viability and modulation of excitatory amino acid transport were verified in hippocampal slices. In-vitro AE or FA was tested against neurotoxicity induced by glutamate or QA. KEY FINDINGS: AE did not prevent QA-induced seizures; however, it prevented cellular death and disruption of excitatory amino acid transport. In-vitro AE (0.1 or 1.0 mg/ml) or FA (1 or 10 µm), improved cell viability against citotoxicity exerted by glutamate or QA, respectively. Both AE and FA have protective effects depending on activation of the phosphatidylinositol-3 kinase (PI3K) signalling pathway. CONCLUSIONS: AE attenuated QA-induced cell damage possibly involving the glutamate transport modulation through NMDAR interaction. FA shows a similar profile of neuroprotection promoted by AE. Therefore, AE treatment might be a useful strategy in preventing brain damage caused by exacerbation of glutamatergic toxicity in nervous system disorders.


Subject(s)
Glutamic Acid/adverse effects , Hippocampus/drug effects , Neurotoxicity Syndromes/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Quinolinic Acid/adverse effects , Verbenaceae/chemistry , Animals , Biological Transport , Cell Death/drug effects , Cell Survival/drug effects , Coumaric Acids/pharmacology , Coumaric Acids/therapeutic use , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acids/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Inbred Strains , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Phosphatidylinositol 3-Kinase/metabolism , Plant Extracts/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Seizures/chemically induced , Seizures/metabolism
18.
J Biol Chem ; 289(14): 9970-82, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24515113

ABSTRACT

The focal swellings of dendrites ("dendritic beading") are an early morphological hallmark of neuronal injury and dendrotoxicity. They are associated with a variety of pathological conditions, including brain ischemia, and cause an acute disruption of synaptic transmission and neuronal network function, which contribute to subsequent neuronal death. Here, we show that increased synaptic activity prior to excitotoxic injury protects, in a transcription-dependent manner, against dendritic beading. Expression of activating transcription factor 3 (ATF3), a nuclear calcium-regulated gene and member of the core gene program for acquired neuroprotection, can protect against dendritic beading. Conversely, knockdown of ATF3 exacerbates dendritic beading. Assessment of neuronal network functions using microelectrode array recordings revealed that hippocampal neurons expressing ATF3 were able to regain their ability for functional synaptic transmission and to participate in coherent neuronal network activity within 48 h after exposure to toxic concentrations of NMDA. Thus, in addition to attenuating cell death, synaptic activity and expression of ATF3 render hippocampal neurons more resistant to acute dendrotoxicity and loss of synapses. Dendroprotection can enhance recovery of neuronal network functions after excitotoxic insults.


Subject(s)
Activating Transcription Factor 3/metabolism , Brain Ischemia/metabolism , Calcium Signaling , Dendrites/genetics , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Transmission , Transcription, Genetic , Activating Transcription Factor 3/genetics , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Death/drug effects , Cell Death/genetics , Dendrites/pathology , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Hippocampus/metabolism , Hippocampus/pathology , Mice , N-Methylaspartate/adverse effects , N-Methylaspartate/pharmacology , Nerve Net/pathology , Nerve Tissue Proteins/genetics
19.
J Clin Psychiatry ; 74(10): 966-73, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24229746

ABSTRACT

OBJECTIVE: Intravenous N-methyl-d-aspartate (NMDA) antagonists have shown promising results in rapidly ameliorating depression symptoms, but placebo-controlled trials of oral NMDA antagonists as monotherapy have not observed efficacy. We conducted a randomized, double-blind, placebo-controlled trial of the NMDA antagonist memantine as an augmentation treatment for patients with DSM-IV major depressive disorder. METHOD: Adult outpatients with major depressive disorder and partial response or nonresponse to their current antidepressant (as indicated by a 17-item Hamilton Depression Rating Scale score of ≥ 16 at baseline) were randomized (from July 2006-December 2011) to add memantine (flexible dose 5-20 mg/d, with all memantine group participants reaching the dose of 20 mg/d) (n = 15) or placebo (n = 16) to their existing treatment for 8 weeks. The primary outcome, change in Montgomery-Asberg Depression Rating Score (MADRS), was evaluated with repeated-measures mixed effects models using last-observation-carried-forward methods. Secondary outcomes included other depression and anxiety rating scales, suicidal and delusional ideation, and other adverse effects. RESULTS: 84% of participants completed the trial, including 93% of participants receiving memantine. Participants receiving memantine did not show a statistically or clinically significant change in MADRS scores compared to placebo, either over the entire study (ß = 0.133, favoring placebo, P = .74) or at study completion (week 8 mean [SD] MADRS score change = -7.13 [6.61] [memantine]; -7.25 [11.14] [placebo]; P = .97). A minimal to small effect size (comparing change to baseline variability) favoring placebo was observed (Cohen d = 0.19). Similarly, no substantial effect sizes favoring memantine nor statistically significant between-group differences were observed on secondary efficacy outcomes. CONCLUSIONS: This trial did not detect significant statistical or effect size differences between memantine and placebo augmentation among nonresponders or poor responders to conventional antidepressants. While the small number of participants is a limitation, this study suggests memantine lacks substantial efficacy as an augmentation treatment for major depressive disorder. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00344682.


Subject(s)
Antidepressive Agents , Depression/drug therapy , Depressive Disorder, Major , Memantine , Adult , Antidepressive Agents/classification , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Diagnostic and Statistical Manual of Mental Disorders , Drug Monitoring , Drug Synergism , Drug Therapy, Combination , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/adverse effects , Female , Humans , Male , Memantine/administration & dosage , Memantine/adverse effects , Psychiatric Status Rating Scales , Receptors, N-Methyl-D-Aspartate/metabolism , Treatment Outcome
20.
Clin Cancer Res ; 19(16): 4446-54, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23833301

ABSTRACT

PURPOSE: To test whether memantine can prevent methotrexate-induced cognitive deficits in a preclinical model. EXPERIMENTAL DESIGN: After noting that methotrexate exposure induces prolonged elevations of the glutamate analog homocysteic acid (HCA) within cerebrospinal fluid, we tested whether intrathecal injection of HCA would produce memory deficits similar to those observed after intrathecal methotrexate. We then tested whether memantine, an antagonist of the N-methyl-d-aspartate (NMDA) subclass of glutamate receptors, could protect animals treated with clinically relevant doses of intrathecal methotrexate against developing memory deficits. Finally, we asked whether memantine affected this pathway beyond inhibiting the NMDA receptor by altering expression of the NMDA receptor or affecting concentrations of HCA or glutamate within the central nervous system. RESULTS: Four intrathecal doses of methotrexate induced deficits in spatial memory, persisting at least one month following the final injection. Intrathecal HCA was sufficient to reproduce this deficit. Concurrent administration of memantine during the period of methotrexate exposure was protective, decreasing the incidence of methotrexate-induced spatial memory deficits from 56% to 20% (P < 0.05). Memantine neither altered expression of NMDA receptors within the hippocampus nor blunted the methotrexate-induced increases in glutamate or HCA. CONCLUSIONS: Excitotoxic glutamate analogs including HCA contribute to cognitive deficits observed after intrathecal methotrexate. Memantine, an NMDA receptor antagonist, reduces the incidence of cognitive deficits in rats treated with intrathecal methotrexate, and may therefore benefit patients with cancer receiving similar treatment.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Methotrexate/adverse effects , Animals , Cognition/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/administration & dosage , Injections, Spinal , Male , Memantine/administration & dosage , Methotrexate/administration & dosage , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...