Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 514
Filter
1.
Neurobiol Learn Mem ; 168: 107159, 2020 02.
Article in English | MEDLINE | ID: mdl-31911198

ABSTRACT

Hypofunction of the NMDA receptor (NMDAr) may underlie cognitive deficits associated with schizophrenia and other psychiatric conditions including working memory (WM) impairments. Given that these deficits link closely to functional outcome, treatments remediating such deficits require identification. NMDAr hypofunction can be modeled via treatment with the antagonist MK-801. Hence, the present study determined whether cholinergic or dopaminergic agonists attenuate MK-801-induced WM deficits in mice. WM was assessed in male C57BL/6 mice trained on an automated 12-arm radial arm maze (RAM) paradigm, wherein rewards were delivered after the first but, not after subsequent entries into WM arms (8/12) and never delivered for entries into reference memory (RM) arms (4/12). Mice were then treated with MK-801 (vehicle or 0.3 mg/kg) and nicotine (vehicle, 0.03 or 0.30 mg/kg) in a cross-over design. After a 2-week washout, mice were then retested with MK-801 and the dopamine D2-family receptor agonist bromocriptine (vehicle, 3 or 10 mg/kg). In both experiments, MK-801 reduced WM span and increased RM and WM error rates. Nicotine did not attenuate these deficits. In contrast, a bromocriptine/MK-801 interaction was observed on WM error rate, where bromocriptine attenuated MK-801 induced deficits without affecting MK-801-induced RM errors. Additionally, bromocriptine produced the main effect of slowing latency to collect rewards. Hence, while NMDAr hypofunction-induced deficits in WM was unaffected by nicotine, it was remediated by treatment with the dopamine D2-family agonist bromocriptine. Future studies should determine whether selective activation of dopamine D2, D3, or D4 receptors remediate this NMDAr hypofunction-induced WM deficit.


Subject(s)
Bromocriptine/administration & dosage , Maze Learning/drug effects , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Nicotine/administration & dosage , Receptors, Dopamine D2/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Dizocilpine Maleate/administration & dosage , Dopamine Agonists/administration & dosage , Male , Mice, Inbred C57BL
2.
Int J Neuropsychopharmacol ; 23(7): 459-468, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32725129

ABSTRACT

BACKGROUND: Systemic administration of noncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonists such as MK-801 is widely used to model psychosis of schizophrenia (SZ). Acute systemic MK-801 in rodents caused an increase of the auditory steady-state responses (ASSRs), the oscillatory neural responses to periodic auditory stimulation, while most studies in patients with SZ reported a decrease of ASSRs. This inconsistency may be attributable to the comprehensive effects of systemic administration of MK-801. Here, we examined how the ASSR is affected by selectively blocking NMDAR in the thalamus. METHODS: We implanted multiple electrodes in the auditory cortex (AC) and prefrontal cortex to simultaneously record the local field potential and spike activity (SA) of multiple sites from awake mice. Click-trains at a 40-Hz repetition rate were used to evoke the ASSR. We compared the mean trial power and phase-locking factor and the firing rate of SA before and after microinjection of MK-801 (1.5 µg) into the medial geniculate body (MGB). RESULTS: We found that both the AC and prefrontal cortex showed a transient local field potential response at the onset of click-train stimulus, which was less affected by the application of MK-801 in the MGB. Following the onset response, the AC also showed a response continuing throughout the stimulus period, corresponding to the ASSR, which was suppressed by the application of MK-801. CONCLUSION: Our data suggest that the MGB is one of the generators of ASSR, and NMDAR hypofunction in the thalamocortical projection may account for the ASSR deficits in SZ.


Subject(s)
Dizocilpine Maleate/pharmacology , Evoked Potentials, Auditory/drug effects , Geniculate Bodies/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Acoustic Stimulation , Animals , Auditory Cortex/drug effects , Dizocilpine Maleate/administration & dosage , Electrodes, Implanted , Electroencephalography , Male , Mice , Mice, Inbred C57BL , Microinjections , Prefrontal Cortex/drug effects , Thalamus/drug effects , Wakefulness
3.
Nutr Neurosci ; 23(9): 672-678, 2020 Sep.
Article in English | MEDLINE | ID: mdl-30465483

ABSTRACT

The study of inbred mouse strains is a useful animal model to assess differences in ingestive behavior responses, including conditioned flavor preferences (CFP). C57BL/6, BALB/c and SWR inbred mice display differential sensitivity to dopamine (DA) D1, opioid and muscarinic cholinergic receptor antagonism of sucrose or saccharin intake as well as to muscarinic cholinergic antagonism of acquisition (learning) of sucrose-CFP. Given that DA D1, opioid and N-methyl-D-aspartate (NMDA) receptor antagonists differentially alter sucrose-CFP in BALB/c and SWR inbred mice, the present study examined whether systemic administration of naltrexone, SCH23390 or MK-801 altered acquisition and expression of sucrose-CFP in C57BL/6 mice. In acquisition experiments, male food-restricted C57BL/6 mice were treated with vehicle, naltrexone (1, 5 mg/kg), SCH23390 (50, 200 nmol/kg) or MK-801 (100, 200 µg/kg) 30 min prior to each of ten daily sessions in which they alternately consumed a flavored (CS+, e.g. cherry) 16% sucrose solution and a differently-flavored (CS-, e.g. grape) 0.05% saccharin solution followed by six two-bottle CS choice tests mixed in 0.2% saccharin without injections. SCH23390 and MK-801, but not naltrexone eliminated sucrose-CFP acquisition in food-restricted C57BL/6 mice. In expression experiments, food-restricted C57BL/6 mice underwent the ten training sessions without injections followed by two-bottle CS choice tests 30 min following vehicle, naltrexone (1, 5 mg/kg), SCH23390 (200, 800 nmol/kg) or MK-801 (100, 200 µg/kg). SCH23390 more effectively reduced the magnitude of sucrose-CFP expression than naltrexone or MK-801 in food-restricted C57BL/6 mice. Thus, dopamine D1 and NMDA receptor signaling is essential for learning of sucrose-CFP in C57BL/6 mice.


Subject(s)
Conditioning, Classical/physiology , Food Preferences/physiology , Receptors, Dopamine D1/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Opioid/physiology , Sucrose/administration & dosage , Animals , Benzazepines/administration & dosage , Conditioning, Classical/drug effects , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists , Food Preferences/drug effects , Male , Mice, Inbred C57BL , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
4.
Drug Dev Ind Pharm ; 46(4): 587-596, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32162981

ABSTRACT

Iloperidone (ILO) is an anti-psychotic, used in schizophrenia. It has low bioavailability (36%) due to low solubility and first pass effect. Oral solid self microemulsifying drug delivery system (SMEDDS) and liquisolid compact (LSC) of ILO were developed. The hypothesis is to test in vivo performance (PK and PD effects) of these delivery systems, as both systems improve dissolution. Based on solubility Capmul MCM, Labrafac WL 1349 were selected as oils, Lauroglycol 90 and PEG 600 were selected as surfactant and cosurfactant. Syloid XDP was optimized for adsorption of liquid SMEDDS. Syloid XDP and Aerosil 200 were optimized as carrier and coating material in the ratio of 15:1 w/w for liquisolid formulation. SEM and PXRD studies indicated no specific crystallinity due to bulkiness in both formulations, which showed similar flow and release behavior. Pharmacokinetic studies were performed for ILO Coarse suspension (CS), Tablet suspension (TS), optimized solid SMEDDS (A1X) and liquisolid compact (S3) in wistar rats. About 3.80 and 2.19-fold improvements in relative bioavailabilty were found for A1X and S3, respectively, when compared to CS. In comparison to TS, 2.61 and 1.51 fold improvements in bioavailability were found for A1X and S3, respectively. Further, Pharmacodynamic activity was studied by reversal of MK-801 induced hyperlocomotion in rats. A1X and S3 formulations showed maximum reversal after 15 min when compared to CS and found to have similar performance. Thus, in comparison to S3, A1X showed significant difference in pharmacokinetic effects but similar pharmacodynamic effects.


Subject(s)
Antipsychotic Agents/administration & dosage , Drug Carriers/chemistry , Isoxazoles/administration & dosage , Locomotion/drug effects , Piperidines/administration & dosage , Administration, Oral , Animals , Antipsychotic Agents/pharmacokinetics , Biological Availability , Chemistry, Pharmaceutical , Dizocilpine Maleate/administration & dosage , Emulsions , Excipients/chemistry , Humans , Isoxazoles/pharmacokinetics , Male , Models, Animal , Particle Size , Piperidines/pharmacokinetics , Rats , Schizophrenia/drug therapy , Solubility , Surface-Active Agents/chemistry , Suspensions , Tablets
5.
Neurobiol Learn Mem ; 161: 57-62, 2019 05.
Article in English | MEDLINE | ID: mdl-30902736

ABSTRACT

NMDA receptor-dependent synaptic plasticity has been proposed to be important for encoding of memories. Consistent with this hypothesis, the non-competitive NMDA receptor antagonist, MK-801, has been found to impair performance on tests of memory. Interpretation of some of these findings has, however, been complicated by the fact that the drug-state of animals has differed during encoding and tests of memory. Therefore, it is possible that MK-801 may result in state-dependent retrieval or expression of memory rather than actually impairing encoding itself. We tested this hypothesis in mice using tests of object recognition memory with a 24-hour delay between the encoding and test phase. Mice received injections of either vehicle or MK-801 prior to the encoding phase and the test phase. In Experiment 1, a low dose of MK-801 (0.01 mg/kg) impaired performance when the drug-state (vehicle or MK-801) of mice changed between encoding and test, but there was no significant effect of MK-801 on encoding. In Experiment 2, a higher dose of MK-801 (0.1 mg/kg) failed to impair object recognition memory when mice received the drug prior to both encoding and test compared to mice that received vehicle. MK-801 did not affect object exploration, but it did induce locomotor hyperactivity at the higher dose. These results suggest that some previous demonstrations of MK-801 effects may reflect a failure to express or retrieve memory due to the state-dependency of memory rather than impaired encoding of memory.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Memory, Long-Term/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Recognition, Psychology/drug effects , Animals , Behavior, Animal/drug effects , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Female , Habituation, Psychophysiologic/drug effects , Mice , Mice, Inbred C57BL
6.
Learn Mem ; 25(4): 158-164, 2018 04.
Article in English | MEDLINE | ID: mdl-29545387

ABSTRACT

NMDA receptors (NMDARs) are considered critical for the consolidation of extinction but recent work challenges this assumption. Namely, NMDARs are not required for extinction retention in infant rats as well as when extinction training occurs for a second time (i.e., reextinction) in adult rats. In this study, a possible third instance of NMDAR-independent extinction was tested. Although adolescents typically exhibit impaired extinction retention, rats that are conditioned as juveniles and then given extinction training as adolescents (JuvCond-AdolesExt) have good extinction retention. Unexpectedly, this good extinction retention is not associated with an up-regulation of a synaptic plasticity marker in the medial prefrontal cortex, a region implicated in extinction consolidation. In the current study, rats received either the noncompetitive NMDAR antagonist MK801 (0.1 mg/kg, s.c.) or saline before extinction training. In several experiments, rats conditioned and extinguished as juveniles, adolescents, or adults exhibited impaired extinction retention after MK801 compared to saline, but this effect was not observed in JuvCond-AdolesExt rats. Further experiments ruled out several alternative explanations for why NMDAR antagonism did not affect extinction retention in adolescents extinguishing fear learned as a juvenile. These results illustrate yet another circumstance in which NMDARs are not required for successful extinction retention and highlight the complexity of fear inhibition across development.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Conditioning, Classical , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Extinction, Psychological/drug effects , Male , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
7.
Cancer Sci ; 109(12): 3874-3882, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30298963

ABSTRACT

Autocrine and paracrine factors, including glutamate and epidermal growth factor (EGF), are potent inducers of brain tumor cell invasion, a pathological hallmark of malignant gliomas. System xc(-) consists of xCT and CD98hc subunits and functions as a plasma membrane antiporter for the uptake of extracellular cystine in exchange for intracellular glutamate. We previously showed that the EGF receptor (EGFR) interacts with xCT and thereby promotes the activity of system xc(-) in a kinase-independent manner, resulting in enhanced glutamate release in glioma cells. However, the molecular mechanism underlying EGFR-mediated glioma progression in a glutamate-rich microenvironment has remained unclear. Here we show that the GluN2B subunit of the N-methyl-d-aspartate-sensitive glutamate receptor (NMDAR) is a substrate of EGFR in glioma cells. In response to EGF stimulation, EGFR phosphorylated the COOH-terminal domain of GluN2B and thereby enhanced glutamate-NMDAR signaling and consequent cell migration in EGFR-overexpressing glioma cells. Treatment with the NMDAR inhibitor MK-801 or the system xc(-) inhibitor sulfasalazine suppressed EGF-elicited glioma cell migration. The administration of sulfasalazine and MK-801 also synergistically suppressed the growth of subcutaneous tumors formed by EGFR-overexpressing glioma cells. Furthermore, shRNA-mediated knockdown of xCT and GluN2B cooperatively prolonged the survival of mice injected intracerebrally with such glioma cells. Our findings thus establish a central role for EGFR in the signaling crosstalk between xCT and GluN2B-containing NMDAR in glioma cells.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Drug Synergism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Glioma/drug therapy , Glutamic Acid/metabolism , Humans , Mice , Neoplasm Transplantation , Phosphorylation , Protein Domains , Receptors, N-Methyl-D-Aspartate/chemistry , Signal Transduction/drug effects , Sulfasalazine/administration & dosage , Sulfasalazine/pharmacology
8.
Neurobiol Learn Mem ; 154: 5-11, 2018 10.
Article in English | MEDLINE | ID: mdl-29438741

ABSTRACT

Metaplasticity is the dynamic regulation of the ability to induce activity-dependent synaptic plasticity and is governed by the prior history of the synapses. Previous reports by others and us have shown that behavioral stress induces a form of emotional metaplasticity that affects the ability to induce LTP in the subiculum-medial prefrontal cortex pathway, which depends on NMDA receptors (NMDAr). However, studies addressing the effects of stress on LTP and metaplasticity have mainly focused on the adult animal. Here we compared the effects of exposure to stress on the induction of LTP in adult and juvenile animals and examined whether a low dose of NMDAr antagonist (MK801) that does not affect LTP per se would differentially affect stress-induced metaplasticity in adult and juvenile animals. Our findings show that exposure to the elevated platform differentially affects the induction of LTP in adult and juvenile animals. Specifically, whereas exposure to stress resulted in impaired LTP in adult animals, it resulted in enhanced LTP in juvenile animals. Similarly, while MK801 failed to inhibit the induction of LTP in both age groups, it resulted in inhibition of stress-induced enhanced LTP in juvenile animals, but did not affect stress-induced impaired LTP in adult animals. Taken together, these findings demonstrate that emotional metaplasticity is differently dependent on NMDAr in adult and juvenile animals that may stem from developmental differences in the NMDA receptor representation. These results further confirm that the mechanisms of plasticity following stress are distinctive in the two groups of age.


Subject(s)
Emotions/physiology , Long-Term Potentiation , Prefrontal Cortex/physiology , Stress, Psychological , Age Factors , Animals , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Long-Term Potentiation/drug effects , Male , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
9.
Neurobiol Learn Mem ; 154: 87-96, 2018 10.
Article in English | MEDLINE | ID: mdl-29397283

ABSTRACT

Metaplasticity, defined as the plasticity of synaptic plasticity, could affect learning and memory at different neural levels. It was hypothesized that metaplasticity changes on glutamate receptors may affect memory destabilization, promoting or preventing reconsolidation. We investigated the metaplastic effect of NMDA channel blocker MK-801 on sucrose instrumental memory reconsolidation in a behavioural rat model associated to the assessment of molecular markers of metaplasticity, memory retrieval, destabilization and reconsolidation. Following instrumental conditioning and forced abstinence, rats were intraperitoneally treated with MK-801 or vehicle 24 h before the exposure to memory retrieval or not-retrieval. Separate groups were tested for in-vivo extinction of responding (24 h and 7 d after reactivation) or ex-vivo assessment of transcription factor Zif268 and ribosomal protein rpS6 phosphorylation in nucleus accumbens (NAc) and amygdala (Amy). MK-801 significantly inhibited instrumental responding at extinction test, suggesting reconsolidation blockade of instrumental memory. The decrease of Zif268 and phosphorylated-rpS6 levels in NAc and Amy in MK-801/Retrieval vs. Vehicle/Retrieval group supported the behavioural findings. An increase of GluN2B, GluA1 and mGluR5 in NAc, and GluN2B in Amy, 24 h after MK-801 indicated the trigger of associated metaplastic changes. Our findings show that metaplastic changes induced by NMDA receptors blockade affected sucrose instrumental memory retrieval as shown by both behavioural and molecular changes. We hypothesize that these findings however suggested a switch to extinction rather than a reconsolidation.


Subject(s)
Brain/physiology , Conditioning, Operant/physiology , Memory Consolidation/physiology , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain/drug effects , Conditioning, Operant/drug effects , Dizocilpine Maleate/administration & dosage , Early Growth Response Protein 1/metabolism , Excitatory Amino Acid Antagonists/administration & dosage , Male , Memory Consolidation/drug effects , Neuronal Plasticity/drug effects , Protein Subunits/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Ribosomal Protein S6/metabolism , Sucrose/administration & dosage
10.
Int J Mol Sci ; 19(11)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463253

ABSTRACT

To explore pathophysiology of schizophrenia, this study analyzed the regulation mechanisms that are associated with cystine/glutamate antiporter (Sxc), group-II (II-mGluR), and group-III (III-mGluR) metabotropic glutamate-receptors in thalamo-cortical glutamatergic transmission of MK801-induced model using dual-probe microdialysis. L-glutamate release in medial pre-frontal cortex (mPFC) was increased by systemic- and local mediodorsal thalamic nucleus (MDTN) administrations of MK801, but was unaffected by local administration into mPFC. Perfusion into mPFC of activators of Sxc, II-mGluR, and III-mGluR, and into the MDTN of activators of Sxc, II-mGluR, and GABAA receptor inhibited MK801-evoked L-glutamate release in mPFC. Perfusion of aripiprazole (APZ) into MDTN and mPFC also inhibited systemic MK801-evoked L-glutamate release in mPFC. Inhibition of II-mGluR in mPFC and MDTN blocked inhibitory effects of Sxc-activator and APZ on MK801-evoked L-glutamate release; however, their inhibitory effects were blocked by the inhibition of III-mGluR in mPFC but not in MDTN. These results indicate that reduced activation of the glutamate/NMDA receptor (NMDAR) in MDTN enhanced L-glutamate release in mPFC possibly through GABAergic disinhibition in MDTN. Furthermore, MDTN-mPFC glutamatergic transmission receives inhibitory regulation of Sxc/II-mGluR/III-mGluR functional complex in mPFC and Sxc/II-mGluR complex in MDTN. Established antipsychotic, APZ inhibits MK801-evoked L-glutamate release through the activation of Sxc/mGluRs functional complexes in both MDTN and mPFC.


Subject(s)
Antiporters/metabolism , Aripiprazole/pharmacology , Dizocilpine Maleate/pharmacology , Glutamic Acid/metabolism , N-Methylaspartate/antagonists & inhibitors , Prefrontal Cortex/physiopathology , Synaptic Transmission/drug effects , Thalamus/physiopathology , Acetylcysteine/pharmacology , Animals , Aripiprazole/administration & dosage , Dizocilpine Maleate/administration & dosage , Male , Models, Biological , Perfusion , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Thalamus/drug effects
11.
Am J Physiol Cell Physiol ; 313(2): C187-C196, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28566490

ABSTRACT

Major depression is an important clinical factor in ventricular arrhythmia. Patients diagnosed with major depression overexpress N-methyl-d-aspartate receptors (NMDARs). Previous studies found that chronic NMDAR activation increases susceptibility to ventricular arrhythmias. We aimed to explore the mechanisms by which NMDAR activation may increase susceptibility to ventricular arrhythmias. Male rats were randomly assigned to either normal environments as control (CTL) group or 4 wk of chronic mild stress (CMS) to produce a major depression disorder (MDD) model group. After 4 wk of CMS, depression-like behaviors were measured in both groups. Varying doses (1-100 µM) of NMDA and 10 µM NMDA antagonist (MK-801) were perfused through ventricular myocytes isolated from MDD rats to measure the L-type calcium current (ICa-L) and transient outward potassium current (Ito). Structural remodeling was assessed using serial histopathology including Masson's trichrome dye. Electrophysiological characteristics were evaluated using Langendorff perfusion. Depression-like behaviors were observed in MDD rats. MDD rats showed longer action potential durations at 90% repolarization and higher susceptibility to ventricular arrhythmias than CTL rats. MDD rats showed lower ICa-L and Ito current densities than CTL rats. Additionally, NMDA reduced both currents in a concentration-dependent manner, whereas there was no significant impact on the currents when perfused with MK-801. MDD rats exhibited significantly more fibrosis areas in heart tissue and reduced expression of Kv4.2, Kv4.3, and Cav1.2. We observed that acute NMDAR activation led to downregulation of potassium and L-type calcium currents in a rat model of depression, which may be the mechanism underlying ventricular arrhythmia promotion by depression.


Subject(s)
Calcium Channels, L-Type/genetics , Depressive Disorder, Major/genetics , Myocytes, Cardiac/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Shal Potassium Channels/genetics , Action Potentials/drug effects , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Disease Models, Animal , Dizocilpine Maleate/administration & dosage , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Myocytes, Cardiac/drug effects , N-Methylaspartate/administration & dosage , Patch-Clamp Techniques , Potassium/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Shal Potassium Channels/metabolism
12.
J Neurophysiol ; 118(2): 1002-1011, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28539393

ABSTRACT

Symptoms of schizophrenia have been linked to insults during neurodevelopment such as NMDA receptor (NMDAR) antagonist exposure. In animal models, this leads to schizophrenia-like behavioral symptoms as well as molecular and functional changes within hippocampal and prefrontal regions. The aim of this study was to determine how administration of the NMDAR antagonist phencyclidine (PCP) during neurodevelopment affects functional network activity within the hippocampus and medial prefrontal cortex (mPFC). We recorded field potentials in vivo after electrical brain stem stimulation and observed a suppression of evoked theta power in ventral hippocampus, while evoked gamma power in mPFC was enhanced in rats administered with PCP neonatally. In addition, increased gamma synchrony elicited by acute administration of the NMDAR antagonist MK-801 was exaggerated in neonatal PCP animals. These data suggest that NMDAR antagonist exposure during brain development alters functional networks within hippocampus and mPFC possibly contributing to the reported behavioral symptoms of this animal model of schizophrenia.NEW & NOTEWORTHY We show that insults with a NMDA receptor antagonist during neurodevelopment lead to suppressed evoked theta oscillations in ventral hippocampus in adult rats, while evoked gamma oscillations are enhanced and hypersensitive to an acute challenge with a NMDA receptor antagonist in prefrontal cortex. These observations reveal the significance of neurodevelopmental disturbances in the evolvement of schizophrenia-like symptoms and contribute to the understanding of the functional deficits underlying aberrant behavior in this disease.


Subject(s)
Disease Models, Animal , Excitatory Amino Acid Antagonists/toxicity , Hippocampus/growth & development , Phencyclidine/toxicity , Prefrontal Cortex/growth & development , Schizophrenia/physiopathology , Animals , Animals, Newborn , Cortical Synchronization/drug effects , Cortical Synchronization/physiology , Dizocilpine Maleate/administration & dosage , Electric Stimulation , Excitatory Amino Acid Antagonists/administration & dosage , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/physiopathology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Random Allocation , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Theta Rhythm/drug effects , Theta Rhythm/physiology
13.
Mol Pain ; 13: 1744806917746564, 2017.
Article in English | MEDLINE | ID: mdl-29166839

ABSTRACT

Triptolide (T10), an active component of Tripterygium wilfordii Hook F, is reported to have potent anti-inflammatory and analgesic effects. Additionally, MK-801, a noncompetitive N-methyl-D-aspartate receptor antagonist, can reduce glutamate toxicity and has a significant analgesic effect on chronic pain. In this study, we tested the possible synergistic analgesic ability by intrathecal administration of T10 and MK-801 for the treatment of neuropathic pain. Single T10 (3, 10, or 30 µg/kg), MK-801 (10, 30, or 90 µg/kg), or a combination of them were intrathecally administrated in rats with spinal nerve ligation. We found that single administration of T10 caused a slow-acting but long-term analgesic effect, while single administration of MK-801 caused a fast-acting but short-term effect. Administration of their combination showed obviously synergic analgesia and the 1:3 ratio of T10 to MK-801 reached the peak effect. Furthermore, application of T10 and/or MK-801 significantly inhibited the activation of microglia and astrocyte and phosphorylation of STAT3 and NR2B in the spinal dorsal horn induced by chronic neuropathic pain. Our data suggest that the combination of T10 and MK-801 may be a potentially novel strategy for treatment of neuropathic pain.


Subject(s)
Diterpenes/therapeutic use , Dizocilpine Maleate/therapeutic use , Neuralgia/drug therapy , Phenanthrenes/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Disease Models, Animal , Diterpenes/administration & dosage , Diterpenes/chemistry , Diterpenes/pharmacology , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/chemistry , Dizocilpine Maleate/pharmacology , Drug Synergism , Epoxy Compounds/administration & dosage , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Hyperalgesia/complications , Hyperalgesia/drug therapy , Injections, Spinal , Ligation , Male , Neuralgia/complications , Neuroglia/drug effects , Neuroglia/metabolism , Phenanthrenes/administration & dosage , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , STAT3 Transcription Factor/metabolism , Spinal Nerves/drug effects , Spinal Nerves/pathology
14.
Neurobiol Learn Mem ; 143: 18-26, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27815216

ABSTRACT

Adolescents, both humans and rodents, exhibit a marked impairment in extinction of fear relative to younger and older groups which could be caused by a failure to efficiently recruit NMDA receptors (NMDARs) in adolescence. It is well-established that systemic administration of NMDAR antagonists (e.g., MK801) before extinction training impairs the retention of extinction in adult and juvenile rodents, but it is unknown whether this is also the case for adolescents. Therefore, in the present study we investigated the effect of pharmacologically manipulating the NMDAR on extinction retention in adolescent rats. When extinction retention is typically impaired (i.e., after one session of extinction training) adolescent male rats given d-cycloserine (a partial NMDAR agonist) showed enhanced extinction retention relative to saline-treated animals while animals given MK801 (a non-competitive antagonist) did not exhibit any further impairment of extinction retention relative to the controls. In a further two experiments we demonstrated that when two sessions of extinction training separated by either 4 or 24h intervals were given to adolescent rats, saline-treated animals exhibited good extinction retention and the animals given MK801 before the second session exhibited impaired extinction retention. These findings suggest that extinction in adolescence does not initially involve NMDARs and this is a likely mechanism that contributes to the impaired fear inhibition observed at this age. However, NMDARs appear to be recruited with extended extinction training or after administration of a partial agonist, both of which lead to effective extinction retention.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Retention, Psychology/physiology , Animals , Conditioning, Classical , Cycloserine/administration & dosage , Dizocilpine Maleate/administration & dosage , Male , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
15.
Neurobiol Learn Mem ; 145: 59-66, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28864239

ABSTRACT

Memory retrieval requires coordinated intra- and inter-regional activity in networks of brain structures. Dysfunction of these networks and memory impairment are seen in many psychiatric disorders, but relatively little is known about how memory retrieval and memory failure are represented at the level of local and regional oscillatory activity. To address this question, we measured local field potentials (LFPs) from mice as they explored a novel context, retrieved memories for contextual fear conditioning, and after administration of two amnestic agents: the NMDA receptor antagonist MK-801 and muscarinic acetylcholine receptor antagonist scopolamine (SCOP). LFPs were simultaneously recorded from retrosplenial cortex (RSC), dorsal hippocampus (DH), and anterior cingulate cortex (ACC), which are involved in processing contextual memories, and analyzed for changes in intra-regional power and inter-regional peak coherence of oscillations across multiple frequency bands. Context encoding and memory retrieval sessions yielded similar patterns of changes across all three structures, including decreased delta power and increased theta peak coherence. Baseline effects of MK-801 and SCOP were primarily targeted to gamma oscillations, but in opposite directions. Both drugs also blocked memory retrieval, as indicated by reduced freezing when mice were returned to the conditioning context, but this common behavioral impairment was only associated with power and peak coherence disruptions after MK-801 treatment. These findings point to neural signatures for memory impairment, whose underlying mechanisms may serve as therapeutic targets for related psychiatric disorders.


Subject(s)
Acetylcholine/physiology , Brain/physiology , Glutamic Acid/physiology , Mental Recall/physiology , Synaptic Transmission , Animals , Brain/drug effects , Cholinergic Antagonists/administration & dosage , Conditioning, Classical , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Fear , Male , Mice, Inbred C57BL , Neural Pathways/physiology , Receptors, Muscarinic/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Scopolamine/administration & dosage
16.
Dev Psychobiol ; 59(1): 39-47, 2017 01.
Article in English | MEDLINE | ID: mdl-27473368

ABSTRACT

Plasticity of the axon initial segment (AIS) is a newly discovered type of structural plasticity that regulates cell excitability. AIS plasticity has been reported to happen during normal development of neocortex and also in a few pathological conditions involving disruption of the inhibition/excitation balance. Here we report on the impact of early environmental interventions on structural plasticity of AIS in the mouse neocortex. C57BL/6 mice were raised in standard or enriched environment (EE) from birth up to the time of experiments and were injected with saline or MK-801 [N-Methyl-D-Aspartate (NMDA) receptor antagonist, 1 mg/kg] on postnatal days (P) 6-10. We used Ankyrin G immunoreactivity to mark the AIS of cortical neurons in two sub-regions of frontal cortex (frontal association area, FrA and secondary motor cortex, M2) and in the secondary visual cortex (V2). In 1-month-old mice, the mean AIS length differed between three areas, with the shortest AISs being observed in V2. Postnatal MK-801 or EE led to shortening of AIS only in the frontal areas. However, exposure to EE restored AIS shortening induced by MK-801. Chronic postnatal MK-801 results in structural plasticity of AIS exclusive to the frontal cortex. EE may modify underlying neuronal mechanisms resulting in restoration of AIS length.


Subject(s)
Axon Initial Segment/physiology , Dizocilpine Maleate/pharmacology , Environment , Excitatory Amino Acid Antagonists/pharmacology , Neocortex/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Axon Initial Segment/drug effects , Dizocilpine Maleate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Mice , Mice, Inbred C57BL , Neocortex/drug effects , Neuronal Plasticity/drug effects
17.
Learn Mem ; 23(8): 422-6, 2016 08.
Article in English | MEDLINE | ID: mdl-27421894

ABSTRACT

Here we tested in rats effects of the procognitive drugs modafinil and methylphenidate on post-acquisition performance in an object-location paired-associates learning (PAL) task. Modafinil (32; 64 mg/kg) was without effect, while higher (9 mg/kg) but not lower (4.5 mg/kg) doses of methylphenidate impaired PAL performance. Likewise, higher but not lower doses of amphetamine (0.4; 0.8 mg/kg) and MK-801 (0.08; 0.12 mg/kg) decreased PAL performance. Impaired PAL performance induced by methylphenidate, amphetamine, and MK801 most likely reflects compromised cognitive function, e.g., retrieval of learned paired associates. Our data suggest that stimulant drugs such as methylphenidate and modafinil might not facilitate performance in hippocampus-related cognitive tasks.


Subject(s)
Benzhydryl Compounds/administration & dosage , Central Nervous System Stimulants/administration & dosage , Methylphenidate/administration & dosage , Paired-Associate Learning/drug effects , Amphetamine/administration & dosage , Animals , Conditioning, Operant , Dizocilpine Maleate/administration & dosage , Dose-Response Relationship, Drug , Male , Modafinil , Nootropic Agents/administration & dosage , Rats , Wakefulness-Promoting Agents
18.
Learn Mem ; 23(9): 486-93, 2016 09.
Article in English | MEDLINE | ID: mdl-27531839

ABSTRACT

Destabilization refers to a memory that becomes unstable when reactivated and is susceptible to disruption by amnestic agents. Here we delineated the cellular mechanism underlying the destabilization of drug memory. Mice were conditioned with methamphetamine (MeAM) for 3 d, and drug memory was assessed with a conditioned place preference (CPP) protocol. Anisomycin (ANI) was administered 60 min after the CPP retrieval to disrupt reconsolidation. We found that destabilization of MeAM CPP after the application of ANI was blocked by the N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801 and the NR2B antagonist ifenprodil (IFN) but not by the NR2A antagonist NVP-AAM077 (NVP). In addition, decrease in the phosphorylation of GluR1 at Serine845 (p-GluR1-Ser845), decrease in spine density, and a reduction in the AMPAR/NMDAR ratio in the basolateral amygdala (BLA) were reversed after the MK-801 treatment. The effect of ANI on destabilization was prevented by the protein phosphatase 2B (calcineurin, CaN) inhibitors cyclosporine A (CsA) and FK-506 and the protein phosphatase 1 (PP1) inhibitors calyculin A (CA) and okadaic acid (OA). These results suggest that memory destabilization involves the activation of NR2B-containing NMDARs, which in turn allows the influx of Ca(2+) Increased intracellular Ca(2+) stimulates CaN, leading to the dephosphorylation and inactivation of inhibitor 1 and the activation of PP1. PP1 then dephosphorylates p-GluR1-Ser845 to elicit AMPA receptor (AMPAR) endocytosis and destabilization of the drug memory.


Subject(s)
Amygdala/enzymology , Memory Consolidation/physiology , Methamphetamine/administration & dosage , Phosphoprotein Phosphatases/physiology , Amygdala/drug effects , Animals , Anisomycin/administration & dosage , Calcium Signaling/drug effects , Conditioning, Classical , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dizocilpine Maleate/administration & dosage , Male , Memory Consolidation/drug effects , Mental Recall/drug effects , Mental Recall/physiology , Mice, Inbred C57BL , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Synthesis Inhibitors/administration & dosage , Quinoxalines/administration & dosage , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
19.
BMC Neurosci ; 17: 3, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26754043

ABSTRACT

BACKGROUND: In highly complex social settings, an animal's motivational drive to pursue an object depends not only on the intrinsic properties of the object, but also on whether the decision-making animal perceives an object as being the most desirable among others. Mimetic desire refers to a subject's preference for objects already possessed by another subject. To date, there are no appropriate animal models for studying whether mimetic desire is at play in guiding the decision-making process. Furthermore, the neuropharmacological bases of decision-making processes are not well understood. In this study, we used an animal model (rat) to investigate a novel food-foraging paradigm for decision-making, with or without a mimetic desire paradigm. RESULTS: Faced with the choice of foraging in a competitive environment, rats preferred foraging for the desirable object, indicating the rats' ability for decision-making. Notably, treatment with the non-competitive N-methyl-D-aspartate receptor antagonist MK-801, but not with the dopamine D1 or D2 receptor antagonists, SCH23390 and haloperidol, respectively, suppressed the food foraging preference when there was a competing resident rat in the cage. None of these three antagonists affected the food-foraging preference for palatable food. Moreover, MK-801 and SCH23390, but not haloperidol, were able to abolish the desirable environment effect on standard food-foraging activities in complex social settings. CONCLUSIONS: These results highlight the concept that mimetic desire exerts a powerful influence on food-foraging decision-making in rats and, further, illustrate the various roles of the glutamatergic and dopaminergic systems in mediating these processes.


Subject(s)
Competitive Behavior/physiology , Decision Making/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Benzazepines/administration & dosage , Competitive Behavior/drug effects , Decision Making/drug effects , Dizocilpine Maleate/administration & dosage , Dopamine D2 Receptor Antagonists/administration & dosage , Food Preferences/drug effects , Food Preferences/physiology , Haloperidol/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
20.
Neurobiol Learn Mem ; 131: 61-75, 2016 05.
Article in English | MEDLINE | ID: mdl-26988613

ABSTRACT

The neural correlates of memory have been usually examined considering that memory retrieval and memory expression are interchangeable concepts. However, our studies in the crab Neohelice (Chasmagnathus) granulata and in other memory models have shown that memory expression is not necessary for memory to be re-activated and become labile. In order to examine putative neural correlates of memory in the crab Neohelice, we contrast changes induced by training in both animal's behavior and neuronal responses in the medulla terminalis using in vivo Ca(2+) imaging. Disruption of long-term memory by the amnesic agents MK-801 or scopolamine (5µg/g) blocks the learning-induced changes in the Ca(2+) responses in the medulla terminalis. Conversely, treatments that lead to an unexpressed but persistent memory (weak training protocol or scopolamine 0.1µg/g) do not block these learning-induced neural changes. The present results reveal a set of changes in the neural activity induced by training that correlates with memory persistence but not with the probability of this memory to be expressed in the long-term. In addition, the study constitutes the first in vivo evidence in favor of a role of the medulla terminalis in learning and memory in crustaceans, and provides a physiological evidence indicating that memory persistence and the probability of memory to be expressed might involve separate components of memory traces.


Subject(s)
Behavior, Animal/physiology , Brachyura/physiology , Excitatory Amino Acid Antagonists/pharmacology , Ganglia, Invertebrate/physiology , Memory/physiology , Muscarinic Antagonists/pharmacology , Animals , Behavior, Animal/drug effects , Brachyura/drug effects , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Ganglia, Invertebrate/drug effects , Male , Memory/drug effects , Memory Consolidation/drug effects , Memory Consolidation/physiology , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Mental Recall/drug effects , Mental Recall/physiology , Muscarinic Antagonists/administration & dosage , Scopolamine/administration & dosage , Scopolamine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL