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










Publication year range
1.
Neurobiol Dis ; 153: 105304, 2021 06.
Article in English | MEDLINE | ID: mdl-33621640

ABSTRACT

CDKL5 (cyclin-dependent kinase-like 5) deficiency disorder (CDD) is a severe neurodevelopmental encephalopathy characterized by early-onset epilepsy and intellectual disability. Studies in mouse models have linked CDKL5 deficiency to defects in neuronal maturation and synaptic plasticity, and disruption of the excitatory/inhibitory balance. Interestingly, increased density of both GABAergic synaptic terminals and parvalbumin inhibitory interneurons was recently observed in the primary visual cortex of Cdkl5 knockout (KO) mice, suggesting that excessive GABAergic transmission might contribute to the visual deficits characteristic of CDD. However, the functional relevance of cortical GABAergic circuits abnormalities in these mutant mice has not been investigated so far. Here we examined GABAergic circuits in the perirhinal cortex (PRC) of Cdkl5 KO mice, where we previously observed impaired long-term potentiation (LTP) associated with deficits in novel object recognition (NOR) memory. We found a higher number of GABAergic (VGAT)-immunopositive terminals in the PRC of Cdkl5 KO compared to wild-type mice, suggesting that increased inhibitory transmission might contribute to LTP impairment. Interestingly, while exposure of PRC slices to the GABAA receptor antagonist picrotoxin had no positive effects on LTP in Cdkl5 KO mice, the selective GABAB receptor antagonist CGP55845 restored LTP magnitude, suggesting that exaggerated GABAB receptor-mediated inhibition contributes to LTP impairment in mutants. Moreover, acute in vivo treatment with CGP55845 increased the number of PSD95 positive puncta as well as density and maturation of dendritic spines in PRC, and restored NOR memory in Cdkl5 KO mice. The present data show the efficacy of limiting excessive GABAB receptor-mediated signaling in improving synaptic plasticity and cognition in CDD mice.


Subject(s)
Epileptic Syndromes/metabolism , GABA-B Receptor Antagonists/pharmacology , GABAergic Neurons/metabolism , Long-Term Potentiation/drug effects , Perirhinal Cortex/drug effects , Protein Serine-Threonine Kinases/genetics , Receptors, GABA-B/metabolism , Spasms, Infantile/metabolism , Animals , Disease Models, Animal , Epileptic Syndromes/genetics , GABA-A Receptor Antagonists/pharmacology , Long-Term Potentiation/genetics , Mice , Mice, Knockout , Neuronal Plasticity , Open Field Test , Perirhinal Cortex/metabolism , Phosphinic Acids/pharmacology , Picrotoxin/pharmacology , Propanolamines/pharmacology , Spasms, Infantile/genetics
2.
Neurobiol Learn Mem ; 173: 107264, 2020 09.
Article in English | MEDLINE | ID: mdl-32504664

ABSTRACT

Rats are often reluctant to consume novel tastes because they lack knowledge about the postingestive effects the new foods might have. This paper examines the effect of excitotoxic lesions and temporary inactivation of the perirhinal cortex (Prh), a key region in the recognition memory system, on taste neophobia and its attenuation. Using a two-bottle choice paradigm (saccharin vs water), excitotoxic lesions were found to disrupt taste neophobia to 0.3% and 0.5% saccharin. However, the lesions had no effect when using a concentration of 0.7%, which is qualitatively aversive (expt. 1a-1c). In a second series of experiments the same animals were able to acquire a flavor preference learning on the basis of a flavor-taste association. Lesioned and control rats showed, during a choice test, a clear preference for the flavor associated with saccharin (expt. 2a-2c). Finally, in a third series of experiments, Prh inactivation with lidocaine after trial 1 (expt. 3) and after trials 1-3 (expt. 4) delayed attenuation of the neophobia. These findings suggest that Prh lesions do not significantly affect taste processing/ perception. Prh thus appears to play an essential role in taste neophobia and its attenuation.


Subject(s)
Avoidance Learning/physiology , Perirhinal Cortex/physiology , Recognition, Psychology/physiology , Taste Perception/physiology , Taste/physiology , Animals , Avoidance Learning/drug effects , Male , Perirhinal Cortex/drug effects , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Saccharin/pharmacology , Taste/drug effects , Taste Perception/drug effects
3.
Behav Neurosci ; 134(2): 144-152, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31916795

ABSTRACT

Several studies suggest that the perirhinal cortex (PER) may function to unitize stimulus components across time or modalities. While the PER has been shown to be critical for fear acquisition to discontinuous stimuli, the role of the PER in fear extinction memory has not been evaluated. The current study assessed the involvement of the PER during fear extinction training to a continuous or discontinuous conditioned stimulus (CS). Rats were randomly assigned to 1 of 4 groups based on 2 factors: the CS type (a continuous or discontinuous light) and a pretesting PER manipulation (muscimol inactivation or saline). Results showed that PER inactivation impaired fear memory to both CS types; however, PER inactivation had only impaired extinction memory to the discontinuous light. These results suggest the role of the PER in stimulus unitization extends to supporting the acquisition of fear extinction memory. (PsycINFO Database Record (c) 2020 APA, all rights reserved).


Subject(s)
Conditioning, Classical , Extinction, Psychological/physiology , Fear/physiology , Mental Recall/physiology , Perirhinal Cortex/physiology , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Extinction, Psychological/drug effects , Fear/drug effects , GABA-A Receptor Agonists/administration & dosage , Male , Mental Recall/drug effects , Muscimol/administration & dosage , Perirhinal Cortex/drug effects , Photic Stimulation , Rats, Sprague-Dawley , Visual Perception/drug effects
4.
eNeuro ; 6(6)2019.
Article in English | MEDLINE | ID: mdl-31562178

ABSTRACT

Rett Syndrome is a neurological disorder caused by mutations in the gene encoding methyl CpG binding protein 2 (MeCP2) and characterized by severe intellectual disability. The cholinergic system is a critical modulator of cognitive ability and is affected in patients with Rett Syndrome. To better understand the importance of MeCP2 function in cholinergic neurons, we studied the effect of selective Mecp2 deletion from cholinergic neurons in mice. Mice with Mecp2 deletion from cholinergic neurons were selectively impaired in assays of recognition memory, a cognitive task largely mediated by the perirhinal cortex (PRH). Deletion of Mecp2 from cholinergic neurons resulted in profound alterations in baseline firing of L5/6 neurons and eliminated the responses of these neurons to optogenetic stimulation of cholinergic input to PRH. Both the behavioral and the electrophysiological deficits of cholinergic Mecp2 deletion were rescued by inhibiting ACh breakdown with donepezil treatment.


Subject(s)
Cholinergic Neurons/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Perirhinal Cortex/metabolism , Recognition, Psychology/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cholinergic Neurons/drug effects , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Donepezil/pharmacology , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Optogenetics , Perirhinal Cortex/drug effects , Phenotype , Recognition, Psychology/drug effects , Rett Syndrome/genetics , Rett Syndrome/metabolism
5.
Cell Rep ; 28(3): 640-654.e6, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31315044

ABSTRACT

We remember our lives as sequences of events, but it is unclear how these memories are controlled during retrieval. In rats, the medial prefrontal cortex (mPFC) is positioned to influence sequence memory through extensive top-down inputs to regions heavily interconnected with the hippocampus, notably the nucleus reuniens of the thalamus (RE) and perirhinal cortex (PER). Here, we used an hM4Di synaptic-silencing approach to test our hypothesis that specific mPFC→RE and mPFC→PER projections regulate sequence memory retrieval. First, we found non-overlapping populations of mPFC cells project to RE and PER. Second, suppressing mPFC activity impaired sequence memory. Third, inhibiting mPFC→RE and mPFC→PER pathways effectively abolished sequence memory. Finally, a sequential lag analysis showed that the mPFC→RE pathway contributes to a working memory retrieval strategy, whereas the mPFC→PER pathway supports a temporal context memory retrieval strategy. These findings demonstrate that mPFC→RE and mPFC→PER pathways serve as top-down mechanisms that control distinct sequence memory retrieval strategies.


Subject(s)
Memory, Short-Term/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Receptor, Muscarinic M4/metabolism , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , GABA Antagonists/pharmacology , Hippocampus/physiology , Memory, Short-Term/drug effects , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/physiology , Perirhinal Cortex/drug effects , Perirhinal Cortex/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Rats , Rats, Long-Evans , Receptor, Muscarinic M4/drug effects , Serotonin Antagonists/pharmacology
6.
Neurobiol Learn Mem ; 162: 9-14, 2019 07.
Article in English | MEDLINE | ID: mdl-31047997

ABSTRACT

Nociceptin/Orphanin FQ (N/OFQ) plays an important role in the regulation of spatial, fear and recognition memories. N/OFQ receptors are highly distributed in the perirhinal cortex, which is a key brain area involved in modulating novel object recognition (NOR) memory. However, the role of N/OFQ in NOR memory in the perirhinal cortex was still unknown. Moreover, the effects of N/OFQ on different stages of NOR memory were still unclear. In NOR task, we found that pre-training intracerebroventricular (icv) injection of N/OFQ (0.3 and 1 nmol) impaired long-term memory in a dose-dependent manner. However, icv infusion of N/OFQ immediately after training did not affect NOR memory consolidation even at a high dose of 3 nmol. Pre-test icv injection of N/OFQ (1 nmol) also did not influence NOR memory retrieval. These data indicate that N/OFQ negatively modulates long-term NOR memory during the acquisition phase. Furthermore, the amnesia effect of N/OFQ (1 nmol, icv) could be antagonist by pre-treatment with the selective N/OFQ receptor antagonist [Nphe1]N/OFQ(1-13)NH2 (10 nmol, icv), indicating pharmacological specificity. Then, we found that pre-training infusion of N/OFQ (0.1 and 0.3 nmol/side) into the bilateral perirhinal cortex impaired long-term NOR memory, suggesting the perirhinal cortex is a critical brain structure in mediating the amnesic effect of N/OFQ in NOR task. In conclusion, our data, for the first time, indicate that N/OFQ in the perirhinal cortex impairs NOR memory acquisition through the NOP receptors.


Subject(s)
Memory, Long-Term/drug effects , Opioid Peptides/pharmacology , Perirhinal Cortex/drug effects , Recognition, Psychology/drug effects , Animals , Dose-Response Relationship, Drug , Injections, Intraventricular , Male , Mice , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Nociceptin
7.
Neurobiol Learn Mem ; 162: 36-46, 2019 07.
Article in English | MEDLINE | ID: mdl-31125611

ABSTRACT

In order to optimize outcomes in the face of uncertainty, one must recall past experiences and extrapolate to the future by assigning values to different choice outcomes. This behavior requires an interplay between memory and reward valuation, necessitating communication across many brain regions. At the anatomical nexus of this interplay is the perirhinal cortex (PRC). The PRC is densely connected to the amygdala and orbital frontal cortex, regions that have been implicated in reward-based decision making, as well as the hippocampus. Thus, the PRC could serve as a hub for integrating memory, reward, and prediction. The PRC's role in value-based decision making, however, has not been empirically examined. Therefore, we tested the role of the PRC in a spatial delay discounting task, which allows rats to choose between a 1-s delay for a small food reward and a variable delay for a large food reward, with the delay to the large reward increasing after choice of each large reward and decreasing after each small reward. The rat can therefore adjust the delay by consecutively choosing the same reward or stabilize the delay by alternating between sides. The latter has been shown to occur once the 'temporal cost' of the large reward is established and is a decision-making process termed 'exploitation'. When the PRC was bilaterally inactivated with the GABA(A) agonist muscimol, rats spent fewer trials successfully exploiting to maintain a fixed delay compared to the vehicle control condition. Moreover, PRC inactivation resulted in an increased number of vicarious trial and error (VTE) events at the choice point, where rats had to decide between the two rewards. These behavioral patterns suggest that the PRC is critical for maintaining stability in linking a choice to a reward outcome in the face of a variable cost.


Subject(s)
Choice Behavior/physiology , Delay Discounting/physiology , Perirhinal Cortex/physiology , Spatial Behavior/physiology , Animals , Choice Behavior/drug effects , Delay Discounting/drug effects , GABA-A Receptor Agonists/pharmacology , Male , Muscimol/pharmacology , Perirhinal Cortex/drug effects , Rats , Spatial Behavior/drug effects , Time Factors
8.
Biol Psychiatry ; 86(3): 230-239, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30635130

ABSTRACT

BACKGROUND: A method that promotes the retrieval of lost long-term memories has not been well established. Histamine in the central nervous system is implicated in learning and memory, and treatment with antihistamines impairs learning and memory. Because histamine H3 receptor inverse agonists upregulate histamine release, the inverse agonists may enhance learning and memory. However, whether the inverse agonists promote the retrieval of forgotten long-term memory has not yet been determined. METHODS: Here, we employed multidisciplinary methods, including mouse behavior, calcium imaging, and chemogenetic manipulation, to examine whether and how the histamine H3 receptor inverse agonists, thioperamide and betahistine, promote the retrieval of a forgotten long-term object memory in mice. In addition, we conducted a randomized double-blind, placebo-controlled crossover trial in healthy adult participants to investigate whether betahistine treatment promotes memory retrieval in humans. RESULTS: The treatment of H3 receptor inverse agonists induced the recall of forgotten memories even 1 week and 1 month after training in mice. The memory recovery was mediated by the disinhibition of histamine release in the perirhinal cortex, which activated the histamine H2 receptor. Histamine depolarized perirhinal cortex neurons, enhanced their spontaneous activity, and facilitated the reactivation of behaviorally activated neuronal ensembles. A human clinical trial revealed that treatment of H3 receptor inverse agonists is specifically more effective for items that are more difficult to remember and subjects with poorer performance. CONCLUSIONS: These results highlight a novel interaction between the central histamine signaling and memory engrams.


Subject(s)
Histamine Agonists/pharmacology , Memory Disorders/drug therapy , Mental Recall/drug effects , Perirhinal Cortex/drug effects , Adult , Animals , Betahistine , Cognition/drug effects , Double-Blind Method , Female , Humans , Male , Mice , Mice, Inbred C57BL , Object Attachment , Piperidines , Stochastic Processes , Young Adult
9.
Neurobiol Aging ; 73: 135-144, 2019 01.
Article in English | MEDLINE | ID: mdl-30342274

ABSTRACT

Unitization, the capacity to encode associations as one integrated entity, can enhance associative memory in populations with an associative memory deficit by promoting familiarity-based associative recognition. Patients with Alzheimer's disease (AD) are typically impaired in associative memory compared with healthy controls but do not benefit from unitization strategies. Using fragmented pictures of objects, this study aimed at assessing which of the cognitive processes that compose unitization is actually affected in AD: the retrieval of unitized representations itself, or some earlier stages of processing, such as the integration process at a perceptual or conceptual stage of representation. We also intended to relate patients' object unitization capacity to the integrity of their perirhinal cortex (PrC), as the PrC is thought to underlie unitization and is also one of the first affected regions in AD. We evaluated perceptual integration capacity and subsequent memory for those items that have supposedly been unitized in 23 mild AD patients and 20 controls. We systematically manipulated the level of perceptual integration during encoding by presenting object pictures that were either left intact, separated into 2 fragments, or separated into 4 fragments. Subjects were instructed to unitize the fragments into a single representation. Success of integration was assessed by a question requiring the identification of the object. Participants also underwent a structural magnetic resonance imaging examination, and measures of PrC, posterior cingulate cortex volume and thickness, and hippocampal volume, were extracted. The results showed that patients' perceptual integration performance decreased with the increased fragmentation level and that their memory for unitized representations was impaired whatever the demands in terms of perceptual integration at encoding. Both perceptual integration and memory for unitized representations were related to the integrity of the PrC, and memory for unitized representations was also related to the volume of the hippocampus. We argue that, globally, this supports representational theories of memory that hold that the role of the PrC is not only perceptual nor mnemonic but instead underlies complex object representation.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/psychology , Cognition , Memory , Perception , Perirhinal Cortex/pathology , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Female , Humans , Magnetic Resonance Imaging , Male , Neuroimaging , Perirhinal Cortex/diagnostic imaging , Perirhinal Cortex/drug effects
10.
Pharmacol Biochem Behav ; 175: 89-100, 2018 12.
Article in English | MEDLINE | ID: mdl-30240581

ABSTRACT

Chronic methamphetamine (meth) abuse often turns into a compulsive drug-taking disorder accompanied by persistent cognitive deficits and re-occurring psychosis. Possible common neurobiological substrates underlying meth-induced deficits and schizophrenia remain poorly understood. Serotonin 2A (5-HT2A) and metabotropic glutamate 2 (mGlu2) receptors co-regulate psychosis-like behaviors and cognitive function in animals. Therefore, in the present study we examined the effects of chronic exposure to three different drugs known to produce persistent deficits in sensorimotor gating and cognition [meth, phencyclidine (PCP) and MK-801] on the expression of 5-HT2A and mGlu2 within the rat medial prefrontal cortex (mPFC), dorsal hippocampus (dHPC) and perirhinal cortex (PRh). Adult male rats underwent 14 days of: (a) meth self-administration (6 h/day), (b) phencyclidine (PCP; 5 mg/kg, twice/day) administration, or (c) MK-801 (0.3 mg/kg, twice/day) administration. Seven days after the discontinuation of drug administration, tissues of interest were collected for protein expression analysis. We found that despite different pharmacological mechanism of action, chronic meth, PCP, and MK-801 similarly dysregulated 5-HT2A and mGlu2, as indicated by an increase in the 5-HT2A/mGlu2 expression ratio in the mPFC (all three tested drugs), PRh (meth and PCP), and dHPC (MK-801 only). Complementary changes in G-protein expression (increase in Gαq and decrease in Gαi) were also observed in the mPFC of meth animals. Finally, we found that 5-HT2A/mGlu2 cooperation can be mediated in part by the formation of the receptor heteromer in some, but not all cortical regions. In summary, these data suggest that a shift towards increased availability (and G-protein coupling) of cortical 5-HT2A vs. mGlu2 receptors may represent a common neurobiological mechanism underlying the emergence of psychosis and cognitive deficits observed in subjects with meth use disorder and schizophrenia.


Subject(s)
Dizocilpine Maleate/pharmacology , Methamphetamine/administration & dosage , Perirhinal Cortex/drug effects , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Antibodies/immunology , Immunoprecipitation , Male , Methamphetamine/pharmacology , Perirhinal Cortex/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Long-Evans , Rats, Wistar , Receptor, Serotonin, 5-HT2A/immunology , Reproducibility of Results
11.
Learn Mem ; 25(9): 410-415, 2018 09.
Article in English | MEDLINE | ID: mdl-30115762

ABSTRACT

Prolonged use of methamphetamine (meth) has been associated with episodic memory deficits in humans, and preclinical rat models of meth self-administration indicate the memory deficits are a consequence of meth use. Others have suggested that the meth-induced memory deficits may promote a cyclical pattern of drug use, abstinence, and relapse, although preclinical evidence for this relationship is somewhat lacking. The memory deficits in preclinical models manifest as a loss of novel object recognition (NOR) memory. These deficits occur one to two weeks after cessation of meth use and involve the perirhinal cortex, a parahippocampal region essential to NOR memory. We hypothesized that a loss of perirhinal cortex function contributes to both the NOR memory deficits and increased vulnerability to relapse in a novel-cue reinstatement model. To test this, we attempted to restore NOR memory in meth rats using an excitatory Gq-DREADD in perirhinal neurons. Activation of these neurons not only reversed the meth-induced deficit in NOR memory, but also restored novelty salience in a novel-cue reinstatement model. Thus, perirhinal cortex functionality contributes to both memory deficits in relapse in a long-access model of meth self-administration in rats, and chemogenetic restoration of perirhinal function restores memory and reduces relapse.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Cues , Genetic Techniques , Memory Disorders/chemically induced , Memory Disorders/therapy , Methamphetamine/pharmacology , Perirhinal Cortex/drug effects , Recognition, Psychology/drug effects , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Recurrence
12.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29464195

ABSTRACT

The amygdala is a critical substrate for learning about cues that signal danger. Less is known about its role in processing innocuous or background information. The present study addressed this question using a sensory preconditioning protocol in male rats. In each experiment, rats were exposed to pairings of two innocuous stimuli in stage 1, S2 and S1, and then to pairings of S1 and shock in stage 2. As a consequence of this training, control rats displayed defensive reactions (freezing) when tested with both S2 and S1. The freezing to S2 is a product of two associations formed in training: an S2-S1 association in stage 1 and an S1-shock association in stage 2. We examined the roles of two medial temporal lobe (MTL) structures in consolidation of the S2-S1 association: the perirhinal cortex (PRh) and basolateral complex of the amygdala (BLA). When the S2-S1 association formed in a safe context, its consolidation required neuronal activity in the PRh (but not BLA), including activation of AMPA receptors and MAPK signaling. In contrast, when the S2-S1 association formed in a dangerous context, or when the context was rendered dangerous immediately after the association had formed, its consolidation required neuronal activity in the BLA (but not PRh), including activation of AMPA receptors and MAPK signaling. These roles of the PRh and BLA show that danger changes the way the mammalian brain stores information about innocuous events. They are discussed with respect to danger-induced changes in stimulus processing.


Subject(s)
Association Learning/physiology , Basolateral Nuclear Complex/physiology , Fear/physiology , Memory Consolidation/physiology , Perirhinal Cortex/physiology , Animals , Association Learning/drug effects , Basolateral Nuclear Complex/drug effects , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Memory Consolidation/drug effects , Perirhinal Cortex/drug effects , Random Allocation , Rats, Sprague-Dawley , Receptors, AMPA/metabolism
13.
Hippocampus ; 27(12): 1217-1223, 2017 12.
Article in English | MEDLINE | ID: mdl-28881444

ABSTRACT

The perirhinal cortex (PRh) is a key region downstream of auditory cortex (ACx) that processes familiarity linked mnemonic signaling. In gerbils, ACx-driven EPSPs recorded in PRh neurons are largely shunted by GABAergic inhibition (Kotak et al., 2015, Frontiers in Neural Circuits, 9). To determine whether inhibitory shunting prevents the induction of excitatory long-term potentiation (e-LTP), we stimulated ACx-recipient PRh in a brain slice preparation using theta burst stimulation (TBS). Under control conditions, without GABA blockers, the majority of PRh neurons exhibited long-term depression. A very low concentration of bicuculline increased EPSP amplitude, but under this condition TBS did not significantly increase e-LTP induction. Since PRh synaptic inhibition included a GABAB receptor-mediated component, we added a GABAB receptor antagonist. When both GABAA and GABAB receptors were blocked, TBS reliably induced e-LTP in a majority of PRh neurons. We conclude that GABAergic transmission is a vital mechanism regulating e-LTP induction in the PRh, and may be associated with auditory learning.


Subject(s)
Long-Term Potentiation/physiology , Neural Inhibition/physiology , Perirhinal Cortex/metabolism , Receptors, GABA/metabolism , Animals , Bicuculline/pharmacology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Antagonists/pharmacology , Gerbillinae , Long-Term Potentiation/drug effects , Microelectrodes , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/metabolism , Perirhinal Cortex/drug effects , Tissue Culture Techniques
14.
Neurobiol Learn Mem ; 137: 36-47, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27815215

ABSTRACT

The ability to use information from the physical world to update behavioral strategies is critical for survival across species. The prefrontal cortex (PFC) supports behavioral flexibility; however, exactly how this brain structure interacts with sensory association cortical areas to facilitate the adaptation of response selection remains unknown. Given the role of the perirhinal cortex (PER) in higher-order perception and associative memory, the current study evaluated whether PFC-PER circuits are critical for the ability to perform biconditional object discriminations when the rule for selecting the rewarded object shifted depending on the animal's spatial location in a 2-arm maze. Following acquisition to criterion performance on an object-place paired association task, pharmacological blockade of communication between the PFC and PER significantly disrupted performance. Specifically, the PFC-PER disconnection caused rats to regress to a response bias of selecting an object on a particular side regardless of its identity. Importantly, the PFC-PER disconnection did not interfere with the capacity to perform object-only or location-only discriminations, which do not require the animal to update a response rule across trials. These findings are consistent with a critical role for PFC-PER circuits in rule shifting and the effective updating of a response rule across spatial locations.


Subject(s)
Association Learning/physiology , Executive Function/physiology , Perirhinal Cortex/physiology , Prefrontal Cortex/physiology , Spatial Learning/physiology , Animals , Association Learning/drug effects , Executive Function/drug effects , GABA-A Receptor Agonists/pharmacology , Male , Muscimol/pharmacology , Neural Pathways/drug effects , Neural Pathways/physiology , Perirhinal Cortex/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Inbred F344 , Spatial Learning/drug effects
15.
Front Neural Circuits ; 10: 44, 2016.
Article in English | MEDLINE | ID: mdl-27378860

ABSTRACT

The perirhinal (PER) and entorhinal cortex (EC) receive input from the agranular insular cortex (AiP) and the subcortical lateral amygdala (LA) and the main output area is the hippocampus. Information transfer through the PER/EC network however, is not always guaranteed. It is hypothesized that this network actively regulates the (sub)cortical activity transfer to the hippocampal network and that the inhibitory system is involved in this function. This study determined the recruitment by the AiP and LA afferents in PER/EC network with the use of voltage sensitive dye (VSD) imaging in horizontal mouse brain slices. Electrical stimulation (500 µA) of the AiP induced activity that gradually propagated predominantly in the rostro-caudal direction: from the PER to the lateral EC (LEC). In the presence of 1 µM of the competitive γ-aminobutyric acid (GABAA) receptor antagonist bicuculline, AiP stimulation recruited the medial EC (MEC) as well. In contrast, LA stimulation (500 µA) only induced activity in the deep layers of the PER. In the presence of bicuculline, the initial population activity in the PER propagated further towards the superficial layers and the EC after a delay. The latency of evoked responses decreased with increasing stimulus intensities (50-500 µA) for both the AiP and LA stimuli. The stimulation threshold for evoking responses in the PER/EC network was higher for the LA than for the AiP. This study showed that the extent of the PER/EC network activation depends on release of inhibition. When GABAA dependent inhibition is reduced, both the AiP and the LA activate spatially overlapping regions, although in a distinct spatiotemporal fashion. It is therefore hypothesized that the inhibitory network regulates excitatory activity from both cortical and subcortical areas that has to be transmitted through the PER/EC network.


Subject(s)
Amygdala/physiology , Cerebral Cortex/physiology , Entorhinal Cortex/physiology , Hippocampus/physiology , Inhibition, Psychological , Nerve Net/physiology , Perirhinal Cortex/physiology , Amygdala/drug effects , Animals , Bicuculline/pharmacology , Cerebral Cortex/drug effects , Electric Stimulation , Entorhinal Cortex/drug effects , Female , GABA-A Receptor Antagonists/pharmacology , Male , Mice, Inbred C57BL , Nerve Net/drug effects , Perirhinal Cortex/drug effects , Voltage-Sensitive Dye Imaging
16.
Neurobiol Learn Mem ; 133: 89-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27321161

ABSTRACT

Intra-rhinal cortical infusion of 17-ß estradiol (E2, 244.8pg/µl) enhances performance on the Novel-Object Preference (NOP) test and impairs accuracy on the delayed nonmatching-to-sample (DNMS) task in the same set of ovariectomized rats (Gervais, Jacob, Brake, & Mumby, 2013). These results appear paradoxical, as normal performance on both tests require intact object-recognition memory (ORM) ability. While demonstrating a preference for the novel object requires recognizing the sample object, rodents can recognize the sample object and still fail to demonstrate a preference. Therefore, enhanced NOP test performance is consistent with both improved ORM and increased novel-object exploration independent of memory processes. There is some evidence suggesting that estrogen receptor (ER) ß agonists enhance NOP test performance (Jacome et al., 2010), but no study to date has examined the role of this receptor in DNMS task performance in rodents. The aim of the present study was to determine whether intra-PRh infusion of an ER ß agonist, diarylpropionitrile (DPN, 2µg/µl), has divergent effects on novel-object preference (i.e. novelty preference) and accuracy on the DNMS task. Ovariectomized (OVX) rats (n=7) received chronic low E2 (∼22pg/ml serum) replacement, then intra-PRh infusion of DPN (2µg/µl), E2 (244.8pg/µl), or vehicle before each mixed-delay session (0.5-5min) of the DNMS task. A different set of OVX rats (n=10) received the same infusions before each NOP test trial, and were tested either 4 or 72h later. Consistent with Gervais et al. (2013), intra-PRh E2 reduced accuracy on the DNMS task following a 5-min retention delay and enhanced novelty preference on both tests. Intra-PRh DPN was associated with accuracy that was similar to the vehicle-infusion condition, despite enhancing novelty preference on both tests. The accuracy results suggest that while intra-PRh E2 impairs ORM, ERß does not play a role. However, ERß in the PRh appears to be important for the expression of novelty preference, in a manner that is unaffected by retention delay. These findings suggest that the modulation of novelty preference by intra-PRh E2/ERß may be due to factors unrelated to ORM.


Subject(s)
Behavior, Animal/drug effects , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Perirhinal Cortex/drug effects , Recognition, Psychology/drug effects , Animals , Estradiol/administration & dosage , Estrogens/administration & dosage , Female , Nitriles/administration & dosage , Nitriles/pharmacology , Ovariectomy , Propionates/administration & dosage , Propionates/pharmacology , Rats , Rats, Long-Evans
17.
Hippocampus ; 26(10): 1313-27, 2016 10.
Article in English | MEDLINE | ID: mdl-27258934

ABSTRACT

Although cocaine- and amphetamine-regulated transcript peptide (CART) is detected in several cortical and subcortical areas, its role in higher functions has been largely ignored. We examined the significance of CART in memory formation and tested if the downstream actions of CART involve N-methyl-d-aspartate (NMDA) activated extra-cellular signal-regulated kinase (ERK). Newly formed memory was evaluated using novel object recognition test consisting of familiarization (T1) and choice trials (T2). The choice trials were performed at two time points: 30-min (T230-min ) and 24-h (T224-h ) postacquisition. In choice trial (T230-min ), vehicle control rats explored the novel object for significantly longer duration than the familiar object indicating intact memory formation. However, CART-antibody, U0126 [ERK antagonist, both via intracerebroventricular (icv) or intrahippocampal (ih) route] or MK-801 (NMDA antagonist; intraperitoneal) treated rats spent less time exploring novel objects; CART peptide (icv or ih) was ineffective. During choice trial at T224-h , a significant decrease in novel object exploration time was noticed in vehicle control rats suggesting amnesia. However, treatment with CART, prior to familiarization trial (T1), promoted exploration of the novel object even at T224-h . Pretreatment with U0126 or MK-801 blocked pro-cognitive-like effect of CART suggesting involvement of NMDA-ERK pathway in CART's action. Animals subjected to the object familiarization trial showed a drastic increase in the CART-immunoreactivity in the cells of cornu ammonis 3 and polymorph layer of dentate gyrus, and fibers within ento- (ENT) and peri-rhinal (PRH) cortices. Western blot analysis revealed that CART treatment significantly up-regulated the expression of phospo-ERK1/2 in hippocampus, ENT and PRH. This effect was attenuated following pretreatment with U0126 or MK-801, suggesting the activation of ERK signaling cascade through NMDA receptors. Thus, CART system seems to play an important role in recognition memory and that these effects may be mediated by NMDA receptors-ERK signaling in the ENT/PRH-hippocampal circuit. © 2016 Wiley Periodicals, Inc.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Recognition, Psychology/physiology , Animals , Butadienes/pharmacology , Choice Behavior/drug effects , Choice Behavior/physiology , Cognition/drug effects , Cognition/physiology , Dizocilpine Maleate/pharmacology , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hippocampus/drug effects , Learning/drug effects , Learning/physiology , Male , Nerve Tissue Proteins/administration & dosage , Neurons/drug effects , Neurons/physiology , Neuropsychological Tests , Nitriles/pharmacology , Perirhinal Cortex/drug effects , Perirhinal Cortex/metabolism , Psychotropic Drugs/pharmacology , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Recognition, Psychology/drug effects
18.
Behav Brain Res ; 311: 384-391, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27265784

ABSTRACT

Dysfunctions in the perirhinal cortex (PRh) are associated with visual recognition memory deficit, which is frequently detected in the early stage of Alzheimer's disease. Muscarinic acetylcholine receptor-dependent long-term depression (mAChR-LTD) of synaptic transmission is known as a key pathway in eliciting this type of memory, and Tg2576 mice expressing enhanced levels of Aß oligomers are found to have impaired mAChR-LTD in this brain area at as early as 3 months of age. We found that the administration of Aß oligomers in young normal mice also induced visual recognition memory impairment and perturbed mAChR-LTD in mouse PRh slices. In addition, when mice were treated with infliximab, a monoclonal antibody against TNF-α, visual recognition memory impaired by pre-administered Aß oligomers dramatically improved and the detrimental Aß effect on mAChR-LTD was annulled. Taken together, these findings suggest that Aß-induced inflammation is mediated through TNF-α signaling cascades, disturbing synaptic transmission in the PRh, and leading to visual recognition memory deficits.


Subject(s)
Alzheimer Disease/drug therapy , Infliximab/pharmacology , Memory Disorders/drug therapy , Nootropic Agents/pharmacology , Pattern Recognition, Visual/drug effects , Recognition, Psychology/drug effects , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Disease Models, Animal , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice, Inbred ICR , Pattern Recognition, Visual/physiology , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Perirhinal Cortex/drug effects , Perirhinal Cortex/pathology , Perirhinal Cortex/physiopathology , Receptors, Muscarinic/metabolism , Recognition, Psychology/physiology , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/metabolism
19.
Neurobiol Learn Mem ; 130: 149-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26899993

ABSTRACT

We here explore the utility of a paradigm that allows the simultaneous assessment of memory for object (what) and object location (where) and their comparative predominance. Two identical objects are presented during a familiarity trial; during the test trial one of these is displaced, and a new object is presented in a familiar location. When tested 5 or 80min later, rats explored both the novel and the displaced objects more than two familiar stationary objects, indicating intact memory for both, object and place. When tested 24h later rats explored the novel object more than the displaced familiar one, suggesting that forgetting differently influenced object and place memory, with memory for object being more robust than memory for place. Animals that received post-trial administration of the neurokinin-3 receptor agonist senktide and were tested 24h later, now explored the novel and displaced objects equally, suggesting that the treatment prevented the selective decay of memory for location. Next, animals received NMDA lesions in either the perirhinal cortex or the hippocampus, which are hypothesized to be preferentially involved in memory for objects and memory for place, respectively. When tested 5 or 80min later, the perirhinal cortex lesion group explored the displaced object more, indicating relatively deficient object memory, while the hippocampal lesion led to the opposite pattern, demonstrating comparatively deficient place memory. These results suggest different preferential engagement of the perirhinal cortex and hippocampus in their processing of memory for object and place. This preference test lends itself to application in the comparison of selective lesions of neural sites and projection systems as well as to the assessment of possible preferential action of pharmacological agents on neurochemical processes that subserve object vs place learning.


Subject(s)
Hippocampus/physiology , Memory/physiology , Peptide Fragments/pharmacology , Perirhinal Cortex/physiology , Receptors, Neurokinin-3/agonists , Spatial Behavior/physiology , Substance P/analogs & derivatives , Animals , Hippocampus/drug effects , Male , Memory/drug effects , N-Methylaspartate/toxicity , Perirhinal Cortex/drug effects , Rats , Rats, Wistar , Spatial Behavior/drug effects , Substance P/pharmacology
20.
Neuropsychopharmacology ; 41(6): 1477-85, 2016 May.
Article in English | MEDLINE | ID: mdl-26365953

ABSTRACT

Rats that have self-administered methamphetamine (meth) under long access, but not short access, conditions do not recognize novel objects. The perirhinal cortex is critical for novelty detection, and perirhinal metabotropic glutamate 5 receptors (mGlu5) are downregulated after long-access meth. The novel positive allosteric modulator (PAM) 1-(4-(2,4-difluorophenyl) piperazin-1-yl)-2-((4-fluorobenzyl)oxy)-ethanone, or DPFE, demonstrates improved solubility compared with other mGlu5 PAMs, thus allowing brain-site-specific pharmacological studies. Infusion of DPFE into perirhinal cortex restored novel object recognition in long-access meth rats. To investigate the impact of these cognitive enhancing effects on relapse, we tested the effects of DPFE infusions into perirhinal cortex on meth-seeking under two different test conditions. In the standard cue relapse test, perirhinal DPFE infusions did not alter meth-seeking in the presence of meth cues. However, in a novel cue relapse test, wherein animals were allowed to allocate responding between a novel cue and meth-conditioned cue, perirhinal DPFE infusions shifted the pattern of responding in long-access rats toward a profile resembling short-access rats, which respond equally for novel and meth cues. Perirhinal mGlu5 are thus a promising pharmacological target for the restoration of cognitive function in meth addicts. Targeting these receptors may also reduce relapse, particularly in situations where novel stimuli compete with conditioned stimuli for control over meth seeking.


Subject(s)
Amphetamine-Related Disorders/physiopathology , Exploratory Behavior/physiology , Methamphetamine , Perirhinal Cortex/physiology , Receptor, Metabotropic Glutamate 5/physiology , Amphetamine-Related Disorders/metabolism , Animals , Cues , Exploratory Behavior/drug effects , Male , Perirhinal Cortex/drug effects , Perirhinal Cortex/metabolism , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/drug effects , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...