Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 449
Filter
2.
Biochem Biophys Res Commun ; 558: 175-182, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33932777

ABSTRACT

The structure of the brain is dramatically altered during the critical period. Physiological substances (neurotransmitters, hormones, etc.) in the body fluctuate significantly before and after sexual maturation. Therefore, the effect of chemical exposure on the central nervous system often differs depending on the developmental stage and sex. We aimed to compare the behavioural effects that emerged from the administration of chemicals to mice of different life stages (immature or mature) and different sex (male or female). We administered mice with domoic acid (DA), a marine poison, and ibotenic acid (IA), found in poisonous mushrooms. These excitatory amino acids act as agonists for glutamate and are potent neurotoxins. Interestingly, the behavioural effects of these chemicals were completely different. Following DA administration, we observed memory deficits only in groups of male mice treated at maturity. Following IA administration, we observed deviations in emotional behaviour in groups of male mice treated at both immaturity and maturity. In contrast, few characteristic changes were detected in all groups of females. Our results support the theory that the behavioural effects of chemical administration vary considerably with developmental stages and sex. In conclusion, our findings promote better understanding of individual differences in excitatory chemical-induced neurotoxicity and provide evidence for future risk strategies and treatments.


Subject(s)
Behavior, Animal/drug effects , Ibotenic Acid/toxicity , Kainic Acid/analogs & derivatives , Administration, Oral , Animals , Behavior, Animal/physiology , Brain/drug effects , Brain/growth & development , Brain/physiology , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/toxicity , Female , Ibotenic Acid/administration & dosage , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Male , Marine Toxins/administration & dosage , Marine Toxins/toxicity , Mice , Mice, Inbred C57BL , Neurotoxins/administration & dosage , Neurotoxins/toxicity , Sex Factors , Sexual Maturation/physiology
3.
Schizophr Bull ; 47(6): 1806-1814, 2021 10 21.
Article in English | MEDLINE | ID: mdl-33928393

ABSTRACT

Pomaglumetad methionil (POM), a group 2 metabotropic glutamate receptor (mGluR2/3) agonist, showed promise as a novel antipsychotic in preclinical research but failed to show efficacy in clinical trials, though it has been suggested that it may be effective in certain patient populations, including early in disease patients. We used the methyazoxymethanol acetate (MAM) rat model of schizophrenia to determine whether POM may prevent the development of dopamine (DA) system dysfunction in a model representative of the hyperdopaminergic state thought to underlie psychosis, compared to control (SAL) rats. MAM and SAL rats were administered either POM (3 mg/kg, i.p.), vehicle (1 ml/kg), or no injection during postnatal day (PD) 31-40. In either late adolescence (PD 47-56) or adulthood (PD 83-96), novel object recognition (NOR) was tested, followed by anesthetized in vivo electrophysiological recordings of VTA DA neuron activity or ventral hippocampal (vHPC) pyramidal neuron activity. MAM rats treated with POM demonstrated increased NOR in adulthood compared to no injection MAM rats, but not compared to vehicle-treated MAM rats. POM-treated MAM rats demonstrated normalized DA neuron population activity and vHPC pyramidal neuron activity compared to vehicle and no injection MAM rats in both late adolescence and adulthood. No significant differences were observed across treatment groups in SAL rats. These results suggest that peripubertal mGluR2/3 agonist administration can prevent the emergence of vHPC pyramidal neuron hyperactivity and increased DA neuron population activity in adult MAM rats.


Subject(s)
Amino Acids/pharmacology , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/drug effects , Receptors, Metabotropic Glutamate/agonists , Schizophrenia/drug therapy , Ventral Tegmental Area/drug effects , Age Factors , Amino Acids/administration & dosage , Animals , Disease Models, Animal , Excitatory Amino Acid Agonists/administration & dosage , Male , Methylazoxymethanol Acetate/pharmacology , Neurotoxins/pharmacology , Rats
4.
Physiol Rep ; 9(6): e14774, 2021 03.
Article in English | MEDLINE | ID: mdl-33769694

ABSTRACT

N-methyl-d-aspartate (NMDA) application has conventionally been used to activate spinal networks to induce locomotion in spinalized animals. We recently described an alternative approach in which application of continuous blue light activates channelrhodopsin-2 in vesicular glutamate transporter 2a (vglut2a)-expressing spinal neurons to produce organized, rhythmic locomotor activity in spinally-transected larval zebrafish. This technique arguably enhances research validity, because endogenous glutamate is released into existing synapses instead of activating only a subset of glutamatergic (NMDA) receptors with an exogenous compound. Here, we explored the viability of this approach in the context of using it for longer-term experiments. Fictive swimming was induced through repetitive application of 10-s blue light stimuli to spinalized preparations for up to 60 min at intervals of 1, 3, or 15 min. Locomotor activity was maintained throughout the experimental timecourse, demonstrating the robustness of the system. Although locomotor bursts remained organized into episodes of activity, the number of bursts elicited during each successive stimulus decreased. This was in contrast to NMDA bath application, in which bursts became less episodically organized while the overall number of bursts remained unchanged. The efficacy of the repetitive optogenetic stimulation paradigm was demonstrated through application of exogenous dopamine, which reversibly decreased the number of bursts produced per stimulus compared with untreated preparations. Finally, increasing the stimulus interval to 15 min lessened, but did not eliminate locomotor fatigue from repetitive activation. Altogether, we established repetitive optogenetic stimulation of vglut2a-expressing neurons as a viable alternative to NMDA application for activation of the zebrafish spinal locomotor network.


Subject(s)
Glutamic Acid/physiology , Locomotion/physiology , Motor Neurons/physiology , N-Methylaspartate/physiology , Neurons/physiology , Optogenetics , Spinal Cord/physiology , Animals , Excitatory Amino Acid Agonists/administration & dosage , Fatigue , Locomotion/drug effects , Models, Animal , Motor Neurons/drug effects , N-Methylaspartate/administration & dosage , Neurons/drug effects , Spinal Cord/drug effects , Swimming , Zebrafish
5.
Neuropharmacology ; 189: 108528, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33773999

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) play an essential role in regulating glutamatergic neurotransmission. Recently, pathogenic missense mutations were identified in genes encoding NMDAR subunits; however, their effect on NMDAR activity is often poorly understood. Here, we examined whether three previously identified pathogenic mutations (M641I, A645S, and Y647S) in the M3 domain of the GluN1 subunit affect the receptor's surface delivery, agonist sensitivity, Mg2+ block, and/or inhibition by the FDA-approved NMDAR blocker memantine. When expressed in HEK293 cells, we found reduced surface expression of GluN1-M641I/GluN2A, GluN1-Y647S/GluN2A, and GluN1-Y647S/GluN2B receptors; other mutation-bearing NMDAR combinations, including GluN1/GluN3A receptors, were expressed at normal surface levels. When expressed in rat hippocampal neurons, we consistently found reduced surface expression of the GluN1-M641I and GluN1-Y647S subunits when compared with wild-type GluN1 subunit. At the functional level, we found that GluN1-M641I/GluN2 and GluN1-A645S/GluN2 receptors expressed in HEK293 cells have wild-type EC50 values for both glutamate and glycine; in contrast, GluN1-Y647S/GluN2 receptors do not produce glutamate-induced currents. In the presence of a physiological concentration of Mg2+, we found that GluN1-M641I/GluN2 receptors have a lower memantine IC50 and slower offset kinetics, whereas GluN1-A645S/GluN2 receptors have a higher memantine IC50 and faster offset kinetics when compared to wild-type receptors. Finally, we found that memantine was the most neuroprotective in hippocampal neurons expressing GluN1-M641I subunits, followed by neurons expressing wild-type GluN1 and then GluN1-A645S subunits in an NMDA-induced excitotoxicity assay. These results indicate that specific pathogenic mutations in the M3 domain of the GluN1 subunit differentially affect the trafficking and functional properties of NMDARs.


Subject(s)
Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Mutation/genetics , Nerve Tissue Proteins/genetics , Protein Subunits/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Mutation/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Surface Properties/drug effects
6.
Behav Brain Funct ; 17(1): 1, 2021 Feb 21.
Article in English | MEDLINE | ID: mdl-33612106

ABSTRACT

BACKGROUND: The nucleus accumbens (NAc) plays a principal role in drug reward. It has been reported that metabotropic glutamate receptors (mGlu receptors) play a key role in the rewarding pathway(s). Previous studies have shown the vast allocation of the different types of mGlu receptors, including mGlu8 receptors, in regions that are associated with opioid rewards, such as the NAc. The aim of the present study was to evaluate the role of mGlu8 receptors within the NAc in the acquisition and expression phases of morphine induced conditioned place preference (CPP). Adult male Wistar rats were bilaterally implanted by two cannulas' in the NAc and were evaluated in a CPP paradigm. Selective mGlu8 receptor allosteric agonist (S-3,4-DCPG) was administered at doses of 0.03, 0.3, and 3 µg/0.5 µL saline per side into the NAc on both sides during the 3 days of morphine (5 mg/kg) conditioning (acquisition) phase, or before place preference test, or post-conditioning (expression) phase of morphine-induced CPP. RESULTS: The results revealed that intra-accumbal administration of S-3,4-DCPG (0.3 and 3 µg) markedly decreased the acquisition in a dose-dependent manner but had no effect on expression of morphine-induced CPP. CONCLUSIONS: The findings suggest that activation of mGlu8 receptors in the NAc dose-dependently blocks the establishment of morphine-induced CPP and reduces the rewarding properties of morphine which may be related to the glutamate activity into the NAc and in reward pathway(s). These data suggest that mGlu8 receptor may be involved in conditioned morphine reward.


Subject(s)
Benzoates/pharmacology , Conditioning, Operant/drug effects , Excitatory Amino Acid Agonists/pharmacology , Glycine/analogs & derivatives , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , Benzoates/administration & dosage , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/administration & dosage , Glycine/administration & dosage , Glycine/pharmacology , Male , Microinjections , Motor Activity/drug effects , Neural Pathways/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Wistar , Reward
7.
Toxicology ; 445: 152610, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33027616

ABSTRACT

Perfluoroalkyl acids (PFAAs) are persistent man-made chemicals, ubiquitous in nature and present in human samples. Although restrictions are being introduced, they are still used in industrial processes as well as in consumer products. PFAAs cross the blood-brain-barrier and have been observed to induce adverse neurobehavioural effects in humans and animals as well as adverse effects in neuronal in vitro studies. The sulfonated PFAA perfluorooctane sulfonic acid (PFOS), has been shown to induce excitotoxicity via the N-methyl-D-aspartate receptor (NMDA-R) in cultures of rat cerebellar granule neurons (CGNs). In the present study the aim was to further characterise PFOS-induced toxicity (1-60 µM) in rat CGNs, by examining interactions between PFOS and elements of glutamatergic signalling and excitotoxicity. Effects of the carboxylated PFAA, perfluorooctanoic acid (PFOA, 300-500 µM) on the same endpoints were also examined. During experiments in immature cultures at days in vitro (DIV) 8, PFOS increased both the potency and efficacy of glutamate, whereas in mature cultures at DIV 14 only increased potency was observed. PFOA also increased potency at DIV 14. PFOS-enhanced glutamate toxicity was further antagonised by the competitive NMDA-R antagonist 3-((R)-2-Carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) at DIV 8. At DIV 8, PFOS also induced glutamate release (9-13 fold increase vs DMSO control) after 1-3 and 24 h exposure, whereas for PFOA a large (80 fold) increase was observed, but only after 24 h. PFOS and PFOA both also increased alanine and decreased serine levels after 24 h exposure. In conclusion, our results indicate that PFOS at concentrations relevant in an occupational setting, may be inducing excitotoxicity, and potentiation of glutamate signalling, via an allosteric action on the NMDA-R or by actions on other elements regulating glutamate release or NMDA-R function. Our results further support our previous findings that PFOS and PFOA at equipotent concentrations induce toxicity via different mechanisms of action.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Cerebellum/drug effects , Excitatory Amino Acid Agonists/toxicity , Fluorocarbons/toxicity , Glutamic Acid/toxicity , Neurons/drug effects , Alkanesulfonic Acids/administration & dosage , Animals , Caprylates/administration & dosage , Cattle , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebellum/pathology , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Amino Acid Agonists/administration & dosage , Female , Fluorocarbons/administration & dosage , Glutamic Acid/administration & dosage , Male , Neurons/pathology , Rats , Rats, Wistar
8.
Mar Drugs ; 18(7)2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32708077

ABSTRACT

Emerging literature suggests that after a stroke, the peri-infarct region exhibits dynamic changes in excitability. In rodent stroke models, treatments that enhance excitability in the peri-infarct cerebral cortex promote motor recovery. This increase in cortical excitability and plasticity is opposed by increases in tonic GABAergic inhibition in the peri-infarct zone beginning three days after a stroke in a mouse model. Maintenance of a favorable excitatory-inhibitory balance promoting cerebrocortical excitability could potentially improve recovery. Brevetoxin-2 (PbTx-2) is a voltage-gated sodium channel (VGSC) gating modifier that increases intracellular sodium ([Na+]i), upregulates N-methyl-D-aspartate receptor (NMDAR) channel activity and engages downstream calcium (Ca2+) signaling pathways. In immature cerebrocortical neurons, PbTx-2 promoted neuronal structural plasticity by increasing neurite outgrowth, dendritogenesis and synaptogenesis. We hypothesized that PbTx-2 may promote excitability and structural remodeling in the peri-infarct region, leading to improved functional outcomes following a stroke. We tested this hypothesis using epicortical application of PbTx-2 after a photothrombotic stroke in mice. We show that PbTx-2 enhanced the dendritic arborization and synapse density of cortical layer V pyramidal neurons in the peri-infarct cortex. PbTx-2 also produced a robust improvement of motor recovery. These results suggest a novel pharmacologic approach to mimic activity-dependent recovery from stroke.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Marine Toxins/administration & dosage , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Oxocins/administration & dosage , Thrombotic Stroke/drug therapy , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Injections , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Transgenic , Recovery of Function , Thrombotic Stroke/metabolism , Thrombotic Stroke/pathology , Thrombotic Stroke/physiopathology
9.
Neuropharmacology ; 170: 108016, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32101763

ABSTRACT

Glaucoma, the second leading cause of irreversible blindness worldwide, is characterized by the selective death of retinal ganglion cells (RGCs). The group II metabotropic glutamate receptor (mGluR II) activation has been linked to RGC survival, however, the mechanism by which it promotes neuronal survival remains poorly defined. In the present work, we show that extracellular application of LY341495, an mGluR II antagonist could increase the RGC firing frequency, suggesting that activation of mGluR II by endogenously released glutamate could modulate RGC excitability. LY354740, an mGluR II agonist, significantly decreased RGC excitability and the reduced presynaptic excitatory inputs and post-synaptic Ca2+-permeable currents mediated the LY354740-induced effects. By using a well-characterized in vivo male Sprague-Dawley rat glaucoma model, we further demonstrate that in the early stage of experimental glaucoma, the expression of mGluR II dimer-formed protein was significantly reduced, and pre-activation of mGluR II by intravitreal injection of LY354740 before establishment of the glaucoma model could effectively reduce excitatory inputs, thereby reversing hyperexcitability induced by elevated intraocular pressure. Furthermore, LY354740 could increase the expression level of brain-derived neurotrophic factor in the glaucomatous retinas, further protecting RGCs. Our study indicates that the abnormal expression of mGluR II may accelerate RGC apoptosis in glaucoma, and demonstrates that mGluR II agonist LY354740 can be used as a novel method to counter RGC apoptosis in glaucoma.


Subject(s)
Bridged Bicyclo Compounds/administration & dosage , Disease Models, Animal , Neurons/drug effects , Ocular Hypertension/drug therapy , Receptors, Metabotropic Glutamate/agonists , Retinal Ganglion Cells/drug effects , Animals , Cell Survival/drug effects , Cell Survival/physiology , Excitatory Amino Acid Agonists/administration & dosage , Intravitreal Injections/methods , Male , Neurons/metabolism , Ocular Hypertension/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Retinal Ganglion Cells/metabolism
10.
Respir Physiol Neurobiol ; 277: 103401, 2020 06.
Article in English | MEDLINE | ID: mdl-32036030

ABSTRACT

The Kölliker-Fuse Nucleus (KF) has been widely investigated for its contribution to "inspiratory off-switch" while more recent studies showed that activation of the Parabrachial Nucleus (PBN) shortened expiratory duration. This study used an adult, in vivo, decerebrate rabbit model to delineate the contribution of each site to inspiratory and expiratory duration through sequential block of glutamatergic excitation with the receptor antagonists 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX) and d(-)-2-amino-5-phosphonopentanoic acid (AP5). Glutamatergic disfacilitation caused large increases in inspiratory and expiratory duration and minor decrease in peak phrenic activity (PPA). Hypoxia only partially reversed respiratory rate depression but PPA was increased to >200 % of control. The contribution of PBN activity to inspiratory and expiratory duration was equal while block of the KF affected inspiratory duration more than expiratory. We conclude that in the in vivo preparation respiratory rate greatly depends on PBN/KF activity, which contributes to the "inspiratory on- "and "off-switch", but is of minor importance for the magnitude of phrenic motor output.


Subject(s)
Glutamic Acid/physiology , Kolliker-Fuse Nucleus/physiology , Parabrachial Nucleus/physiology , Respiratory Center/physiology , Respiratory Rate/physiology , Animals , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Female , Kolliker-Fuse Nucleus/drug effects , Male , Microinjections/methods , Parabrachial Nucleus/drug effects , Quinoxalines/administration & dosage , Rabbits , Respiratory Center/drug effects , Respiratory Rate/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage
11.
Neurobiol Learn Mem ; 169: 107170, 2020 03.
Article in English | MEDLINE | ID: mdl-31978551

ABSTRACT

Pavlovian fear conditioning and extinction procedures have long been used to study the regulation of learned fear. The amygdala is vital for the association of cues and fear expression, whereas the medial prefrontal cortex (mPFC) is critical for fear regulation after extinction. The medial orbitofrontal cortex (mOFC) has an extensive connection with the fear circuit. In human studies, emotional regulation disorders, such as obsessive-compulsive disorder and post-traumatic stress disorder, are often linked to an abnormality in the orbitofrontal cortex (OFC). Therefore, in a series of experiments, we examined whether abnormal mOFC activities interfere with the regulation of learned fear. The mOFC of rats was pharmacologically activated with N-methyl-D-aspartate (NMDA) during the acquisition, early consolidation, or retrieval phase of fear extinction. Under mOFC activation, there was a general initial suppression of the fear response followed by the development of nonspecific fear expression. Moreover, pre-extinction activation of the mOFC abolished extinction acquisition, causing an up-shift in the fear response during the retrieval test. Nonetheless, immediate post-extinction activation of the mOFC did not interfere with extinction consolidation. Overall, our results suggested that mOFC activation abolished fear extinction acquisition and interfered with fear expression.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Prefrontal Cortex/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Conditioning, Classical/physiology , Excitatory Amino Acid Agonists/administration & dosage , Extinction, Psychological/drug effects , Fear/drug effects , Male , N-Methylaspartate/administration & dosage , Prefrontal Cortex/drug effects , Rats, Long-Evans
12.
Brain Res ; 1730: 146640, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31891692

ABSTRACT

Globally, over 50 million people are affected by epilepsy, which is characterized by the occurrence of spontaneous recurrent seizures. Almost one-third of the patients show resistance to current anti-epileptic drugs, making the exploration of new molecular targets necessary. An interesting target may be Homer1, due to its diverse roles in epileptogenesis and synaptic plasticity. Indeed, Homer1 regulates group I metabotropic glutamate (mGlu) receptors (i.e. mGlu1 and mGlu5) scaffolding and signaling in neurons. In the present work, using the systemic kainic acid (KA)-induced status epilepticus (SE) model in adult rats, we investigated the mRNA and protein expression patterns of the mGlu5 receptor, Homer1a and Homer1b/c at 10, 80 and 120 days post-SE (i.e. T10, T80 and T120). Epileptogenesis was validated by electrophysiological recordings of seizures via electroencephalography (EEG) monitoring and through upregulation of glial fibrillary acidic protein. At the protein level, the mGlu5 receptor was downregulated in the late latent phase (T10) and the early- and late exponential growth phase (T80 and T120, respectively), which was best observed in the hippocampal CA1 region. At mRNA level, significant downregulation of the mGlu5 receptor was only detected in the late exponential growth phase. Homer1a expression did not change at any investigated time point. Interestingly, Homer1b/c was only downregulated in the late latent phase, a period where spontaneous seizures are extremely rare. Thus, this phase-specific downregulation may be indicative of an endogenous neuroprotective mechanism. In conclusion, these results suggest that Homer1b/c may be an interesting molecular target to prevent epileptogenesis and/or control seizures.


Subject(s)
Hippocampus/metabolism , Homer Scaffolding Proteins/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Status Epilepticus/metabolism , Animals , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Agonists/administration & dosage , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Kainic Acid/administration & dosage , Male , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Status Epilepticus/chemically induced
13.
Int J Neurosci ; 130(6): 551-560, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31680595

ABSTRACT

Objective: Altered glutamatergic neurotransmission has been implicated in the pathogenesis of obsessive-compulsive disorder (OCD). We examined the effects and potential mechanism of glutamate-related drugs on compulsive behavior in quinpirole (QNP)-sensitized rats, to deepen our understanding of the link between OCD and glutamate.Method: This study systematically compared the effects of the partial NMDA agonist D-Cycloserine and the NMDA antagonist NVP-AAMO77, Ro25-6981 on compulsive behavior using the elevated zero maze, open field, and marble burying tests in QNP-induced OCD model.Results: The competitive N-methyl-D-aspartate glutamate receptor (NMDAR) antagonists NVP-AAMO77 (5 mg/kg) and Ro25-6981 (5 mg/kg) significantly inhibited anxiety-like and compulsive behavior in rats. And D-Cycloserine at all doses showed significant suppression on anxiety-like and marble-burying behavior. Glutamic acid (Glu) levels, reflecting changes in the glutamatergic neurotransmission, were significantly decreased in rat hippocampus of the NVP-AAMO77 and D-Cycloserine-treated group compared to the saline-treated group. The levels of other amino acids were unaffected. Moreover, NVP-AAMO77 significantly decreased the expression of the subunit NR2A of the NMDAR, and Ro25-6981 suppressed the level of the subunit NR2B of the NMDAR, while D-Cycloserine decreased both the subunit NR2A and NR2B of the NMDAR.Conclusion: Collectively, these findings suggest a functional role of NMDARs in anxiety and compulsive behaviors, with NMDARs inhibition promoting anxiolytic-like and anti-compulsive responses. These findings suggest that D-cycloserine, NVP-AAMO77, and Ro25-6981 could be useful drugs for the treatment of OCD, which may be due to the suppression of NR2A- or NR2B- containing NMDAR.


Subject(s)
Anxiety/physiopathology , Compulsive Behavior/physiopathology , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Glutamic Acid/physiology , Obsessive-Compulsive Disorder/physiopathology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
14.
J Integr Neurosci ; 18(2): 163-172, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31321957

ABSTRACT

In this paper, the modulation of ascending commissural interneurons by N-methyl-D-aspartate was investigated in neonatal rats by using retrograde labeling and whole-cell patch clamp. Data shows these interneurons can be divided into three types (single spike, phasic, and tonic) based on their firing patterns. A hyperpolarization-activated nonselective cation current and persistent inward current are expressed in these interneurons. The parameters studied (n = 48) include: resting membrane potential (-59.2 ± 0.8 mV), input resistance (964.4 ± 49.3 MΩ), voltage threshold (-39.5 ± 0.6 mV), rheobase (13.5 ± 0.7 pA), action potential height (55.6 ± 2.2 mV), action potential half-width (2.8 ± 0.1 ms), afterhyperpolarization magnitude (16.1 ± 1.2 mV) and half-decay (217.9 ± 10.7 ms). 10 µM N-methyl-D-aspartate increases excitability of ascending commissural interneurons by depolarizing the membrane potential, hyperpolarizing voltage threshold, reducing rheobase, and shifting the frequency-current relationship to the left. N-methyl-Daspartate enhances persistent inward currents but reduces hyperpolarization-activated nonselective cation currents. This research uncovers unique ionic and intrinsic properties of ascending commissural interneurons which can be modulated by major excitatory neurotransmitters such as N-methyl-D-aspartate to potentially facilitate left-right alternation during locomotion.


Subject(s)
Commissural Interneurons/physiology , Membrane Potentials , N-Methylaspartate/physiology , Spinal Cord/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Commissural Interneurons/cytology , Commissural Interneurons/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Membrane Potentials/drug effects , N-Methylaspartate/administration & dosage , Rats, Wistar , Spinal Cord/cytology , Spinal Cord/drug effects
15.
Neurobiol Learn Mem ; 164: 107043, 2019 10.
Article in English | MEDLINE | ID: mdl-31325496

ABSTRACT

Taste memory recognition is crucial for species survival; thus, the acquisition of conditioned taste aversion (CTA) protects animals against consuming poisons or toxins. In nature, food and poison are confined in the same edible item; however, in the laboratory these food constituents are usually presented separately for experimental analysis. The taste, or conditioned stimulus (CS), can be hours apart from the gastric malaise, or unconditioned stimulus (US); this extended inter-stimulus interval (ISI) allows the analysis of a particular learning phase. Evidence indicates a relevant function of glutamatergic activity in the insular cortex (IC) throughout the ISI. N-methyl-D-aspartate receptors (NMDAR) are crucial during CTA acquisition and retrieval. However, the exact participation of NMDAR in the IC during the ISI has not been demonstrated. Thus, the aim of this work was to evaluate the effects of temporal NMDAR activation during four time frames throughout the ISI of conditioned sugar aversion with bilateral injections of NMDA at a physiological dose (1 µg/µl) in the IC, given (1) immediately before or (2) immediately after sugar presentation, or (3) immediately before or (4) immediately after LiCl i.p. injection. The results showed that NMDAR activation in the IC had a specific ISI effect during CTA acquisition, increasing aversive memory formation and delaying extinction only after CS presentation. Overall, these results demonstrate that NMDAR in the IC have a particular enhancing associative effect after CS and suggest that there is a precise coincidence in neurochemical events in the IC that correlates with the stimulus to be associated and the glutamate NMDAR activity that must be finely tuned in the ISI during CTA acquisition.


Subject(s)
Avoidance Learning/physiology , Cerebral Cortex/physiology , Conditioning, Classical/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Taste Perception/physiology , Animals , Avoidance Learning/drug effects , Cerebral Cortex/drug effects , Conditioning, Classical/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , N-Methylaspartate/administration & dosage , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Time Factors
16.
Pharmacol Biochem Behav ; 183: 72-79, 2019 08.
Article in English | MEDLINE | ID: mdl-31202811

ABSTRACT

One of most formidable problems in the treatment of addiction is the high rate of relapse. The discovery of medicines to help mitigate relapse are aided by animal models that currently involve weeks of training and require surgical preparations and drug delivery devices. The present set of experiments was initiated to investigate a rapid 8-day screening method that utilizes food instead of intravenous drug administration. Male Sprague-Dawley rats were trained in a reinstatement paradigm in which every lever press produced a 45 mg food pellet concurrently paired with a light and tone. Behavior was subsequently extinguished with lever responses producing neither food nor food-associated stimuli. Reinstatement of responding was evaluated under conditions in which the first three responses of every 5 min time bin produced a food pellet along with food-associated stimuli. The mGlu5 receptor antagonists MPEP and MTEP produced a significant reduction in reinstatement while failing to alter responding where every response produced food. The cannabinoid CB1 receptor antagonist rimonabant and the mGlu2/3 receptor agonist LY379268 also selectively reduced reinstatement. Other compounds including clozapine, d-amphetamine, chlordiazepoxide, ABT-431, naltrexone and citalopram were without effect. The results suggest that relapse-like behavioral effects can be extended to non-pharmacological reinforcers. Drug effects demonstrated both behavioral and pharmacological specificity. The present experimental design thus allows for efficient and rapid assessment of the effects of drugs that might be useful in the treatment of addiction-associated relapse.


Subject(s)
Behavior, Addictive/drug therapy , Behavior, Addictive/psychology , Behavior, Animal/drug effects , Drug Discovery/methods , Models, Animal , Amino Acids/administration & dosage , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cannabinoid Receptor Antagonists/administration & dosage , Cannabinoid Receptor Antagonists/pharmacology , Conditioning, Operant/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/drug effects , Feeding Behavior/drug effects , Male , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Recurrence , Rimonabant/administration & dosage , Rimonabant/pharmacology , Self Administration , Substance-Related Disorders/drug therapy , Thiazoles/administration & dosage , Thiazoles/pharmacology
17.
Int J Mol Sci ; 20(11)2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31174329

ABSTRACT

Negative and cognitive symptoms of schizophrenia contribute to an impaired social and professional life for schizophrenic patients, and in most cases, these symptoms are treatment resistant. Therefore, identification of new treatment strategies is sorely needed. Metabotropic glutamate receptors (mGlu) and muscarinic (M) receptors for acetylcholine have been considered promising targets for novel antipsychotics. Among them, mGlu2 and M4 subtypes seem to be of particular importance. In the present study, the effect of mutual activation of mGlu2 and M4 receptors was assessed in MK-801-based animal models of negative and cognitive symptoms of schizophrenia, that is, social interaction and novel object recognition tests. Low sub-effective doses of LY487379 (0.5 mg/kg), a positive allosteric activator of the mGlu2 receptor, and VU152100 (0.25-0.5 mg/kg), a positive allosteric modulator of the M4 receptor, were simultaneously administered in the aforementioned tests. Combined administration of these compounds prevented MK-801-induced disturbances in social interactions and object recognition when acutely administered 30 min before MK-801. Prolonged (7 days) administration of these compounds resulted in the loss of effectiveness in preventing MK-801-induced disruptions in the novel object recognition test but not in the social interaction test. In the next set of experiments, MK-801 (0.3 mg/kg) was administered for seven consecutive days, and the activity of the compounds was investigated on day eight, during which time MK-801 was not administered. In this model, based on prolonged MK-801 administration, the effectiveness of the compounds to treat MK-801-induced disruptions was evident at low doses which were ineffective in preventing the behavioural disturbances induced by an acute MK-801 injection. Combined administration of the compounds did not exert better efficacy than each compound given alone. Pharmacokinetic analysis confirmed a lack of possible drug-drug interactions after combined administration of LY487379 and VU152100. Our data show that modulation of M4 and mGlu2 receptors may potentially be beneficial in the treatment of negative and cognitive symptoms of schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Benzamides/therapeutic use , Memory, Short-Term/drug effects , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Schizophrenia/drug therapy , Social Behavior , Sulfonamides/therapeutic use , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Dizocilpine Maleate/toxicity , Drug Therapy, Combination , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Excitatory Amino Acid Antagonists/toxicity , Male , Mice , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Receptor, Muscarinic M4/drug effects , Receptors, Glutamate , Schizophrenia/etiology , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
18.
Pharmacol Res Perspect ; 7(3): e00479, 2019 06.
Article in English | MEDLINE | ID: mdl-31086673

ABSTRACT

The downregulation of the glutamate system may be involved in positive, negative, and cognitive symptoms of schizophrenia. Through enhanced glutamate signaling, the activation of the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor, an ionotropic glutamate receptor, could be a new therapeutic strategy for schizophrenia. TAK-137 is a novel AMPA receptor potentiator with minimal agonistic activity; in this study, we used rodents and nonhuman primates to assess its potential as a drug for schizophrenia. At 10 mg kg-1 p.o., TAK-137 partially inhibited methamphetamine-induced hyperlocomotion in rats, and at 3, 10, and 30 mg kg-1 p.o., TAK-137 partially inhibited MK-801-induced hyperlocomotion in mice, suggesting weak effects on the positive symptoms of schizophrenia. At 0.1 and 0.3 mg kg-1 p.o., TAK-137 significantly ameliorated MK-801-induced deficits in the social interaction of rats, demonstrating potential improvement of impaired social functioning, which is a negative symptom of schizophrenia. The effects of TAK-137 were evaluated on multiple cognitive domains-attention, working memory, and cognitive flexibility. TAK-137 enhanced attention in the five-choice serial reaction time task in rats at 0.2 mg kg-1 p.o., and improved working memory both in rats and monkeys: 0.2 and 0.6 mg kg-1 p.o. ameliorated MK-801-induced deficits in the radial arm maze test in rats, and 0.1 mg kg-1 p.o. improved the performance of ketamine-treated monkeys in the delayed matching-to-sample task. At 0.1 and 1 mg kg-1 p.o., TAK-137 improved the cognitive flexibility of subchronic phencyclidine-treated rats in the reversal learning test. Thus, TAK-137-type AMPA receptor potentiators with low intrinsic activity may offer new therapies for schizophrenia.


Subject(s)
Excitatory Amino Acid Agonists/administration & dosage , Receptors, AMPA/metabolism , Schizophrenia/drug therapy , Thiadiazines/administration & dosage , Animals , Cognition/drug effects , Disease Models, Animal , Dizocilpine Maleate/adverse effects , Drug Evaluation, Preclinical , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Haplorhini , Male , Maze Learning/drug effects , Methamphetamine/adverse effects , Mice , Phencyclidine , Rats , Schizophrenia/chemically induced , Thiadiazines/chemistry , Thiadiazines/pharmacology
19.
Behav Brain Res ; 367: 149-157, 2019 07 23.
Article in English | MEDLINE | ID: mdl-30940513

ABSTRACT

The ventromedial prefrontal cortex (vmPFC) of rats has reciprocal connections with the gustatory and the hedonic impact coding structures. The main goal of the present study was to investigate the involvement of local neurons of vmPFC and their catecholaminergic innervations in taste preference and taste reactivity test. Therefore, kainate or 6-hydroxydopamine (6-OHDA) lesions were performed in the vmPFC by iontophoretic method. In the first experiment, taste preference was tested to 250 mM and 500 mM glucose solutions over water in two-bottle choice test. In the second experiment, taste reactivity was examined to 4 concentrations of glucose solutions (250 mM, 500 mM, 750 mM and 1000 mM) and 4 concentrations of quinine solutions (0.125 mM, 0.25 mM, 1.25 mM and 2.5 mM). Our results showed, that kainate microlesion of vmPFC did not modify the preference of 250 mM and 500 mM glucose solutions in two-bottle choice test. In contrast, 6-OHDA microlesion of vmPFC resulted in increased preference to the higher concentration of glucose (500 mM) solution over water. Results of taste reactivity test showed that kainate lesion resulted in more ingestive and less rejective responses to 750 mM glucose solution and elevated rejectivity to the higher concentrations (1.25 mM and 2.5 mM) of quinine solutions. 6-OHDA lesion of vmPFC increased the number of ingestive responses to highly concentrated (500 mM, 750 mM and 1000 mM) glucose solutions and decreased the number of ingestive responses to the lower concentration (0.125 mM) of quinine solution. The present data provide evidence for the important role of vmPFC neurons and catecholaminergic innervation of the vmPFC in the regulation of hedonic evaluation of tastes and in the hedonic consummatory behavior.


Subject(s)
Adrenergic Agents/pharmacology , Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/pharmacology , Food Preferences/drug effects , Pleasure/drug effects , Prefrontal Cortex/drug effects , Taste Perception/drug effects , Adrenergic Agents/administration & dosage , Animals , Excitatory Amino Acid Agonists/administration & dosage , Kainic Acid/pharmacology , Male , Oxidopamine/pharmacology , Rats , Rats, Wistar
20.
Neuroscience ; 406: 389-404, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30926548

ABSTRACT

The over-activation of N-methyl-D-aspartate receptors (NMDARs) is the main cause of neuronal death in brain ischemia. Both the NMDAR and the Acid-sensing ion channel 1a (ASIC1a) are present in the postsynaptic membrane of the central nervous system (CNS) and participate in physiological and pathological processes. However, the specific role played by ASIC1a in these processes remains elusive. We hypothesize that NMDARs are the primary mediators of normal synaptic transmission and excitatory neuronal death, while ASIC1a plays a modulatory role in facilitating NMDAR function. Using various experimental approaches including patch-clamp recordings on hippocampal slices and CHO cells, primary cultures of hippocampal neurons, calcium imaging, Western blot, cDNA transfection studies, and transient middle cerebral artery occlusion (tMCAO) mouse models, we demonstrate that stimulation of ASIC1a facilitates NMDAR function and inhibition of ASIC1a suppresses NMDAR over-activation. One of our key findings is that activation of ASIC1a selectively facilitates the NR1/NR2A/NR2B triheteromeric subtype of NMDAR currents. In accordance, inhibition of ASIC1a profoundly reduced the NMDAR-mediated EPSCs in older mouse brains, which are known to express much higher levels of triheteromeric NMDARs than younger brains. Furthermore, brain infarct sizes were reduced by a greater degree in older mice compared to younger ones when ASIC1a activity was suppressed. These data suggest that ASIC1a activity selectively enhances the function of triheteromeric NMDARs and exacerbates ischemic neuronal death especially in older animal brains. We propose ASIC1a as a novel therapeutic target for preventing and reducing the detrimental effect of brain ischemia in humans.


Subject(s)
Acid Sensing Ion Channel Blockers/administration & dosage , Acid Sensing Ion Channels/physiology , Excitatory Amino Acid Agonists/administration & dosage , Nerve Tissue Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Female , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/agonists , Organ Culture Techniques , Receptors, N-Methyl-D-Aspartate/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...