Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 108: 324-338, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28844789

ABSTRACT

Approximately one in 45 children have been diagnosed with Autism Spectrum Disorder (ASD), which is characterized by social/communication impairments. Recent studies have linked a subset of familial ASD to mutations in the Protocadherin 10 (Pcdh10) gene. Additionally, Pcdh10's expression pattern, as well as its known role within protein networks, implicates the gene in ASD. Subsequently, the neurobiology of mice heterozygous for Pcdh10 (Pcdh10+/-) has been investigated as a proxy for ASD. Male Pcdh10+/- mice have demonstrated sex-specific deficits in social behavior, recapitulating the gender bias observed in ASD. Furthermore, in vitro slice preparations of these Pcdh10+/- mice demonstrate selective decreases to high frequency electrophysiological responses, mimicking clinical observations. The direct in vivo ramifications of such decreased in vitro high frequency responses are unclear. As such, Pcdh10+/- mice and their wild-type (WT) littermates underwent in vivo electrocorticography (ECoG), as well as ex vivo amino acid concentration quantification using High Performance Liquid Chromatography (HPLC). Similar to the previously observed reductions to in vitro high frequency electrophysiological responses in Pcdh10+/- mice, male Pcdh10+/- mice exhibited reduced gamma-band (30-80Hz), but not lower frequency (10 and 20Hz), auditory steady state responses (ASSR). In addition, male Pcdh10+/- mice exhibited decreased signal-to-noise-ratio (SNR) for high gamma-band (60-100Hz) activity. These gamma-band perturbations for both ASSR and SNR were not observed in females. Administration of a GABAB agonist remediated these electrophysiological alterations among male Pcdh10+/-mice. Pcdh10+/- mice demonstrated increased concentrations of GABA and glutamine. Of note, a correlation of auditory gamma-band responses with underlying GABA concentrations was observed in WT mice. This correlation was not present in Pcdh10+/- mice. This study demonstrates the role of Pcdh10 in the regulation of excitatory-inhibitory balance as a function of GABA in ASD.


Subject(s)
Baclofen/pharmacology , Cadherins/metabolism , GABA-B Receptor Agonists/pharmacology , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , gamma-Aminobutyric Acid/metabolism , Acoustic Stimulation , Animals , Auditory Perception/drug effects , Auditory Perception/physiology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Cadherins/genetics , Chromatography, High Pressure Liquid , Electrocorticography , Electrodes, Implanted , Evoked Potentials/drug effects , Evoked Potentials/physiology , Female , Glutamine/metabolism , Male , Mice, Transgenic , Protocadherins , Sex Characteristics , Theta Rhythm/drug effects , Theta Rhythm/physiology
2.
Biol Psychiatry ; 81(3): 193-202, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27567313

ABSTRACT

BACKGROUND: Behavioral symptoms in individuals with autism spectrum disorder (ASD) have been attributed to abnormal neuronal connectivity, but the molecular bases of these behavioral and brain phenotypes are largely unknown. Human genetic studies have implicated PCDH10, a member of the δ2 subfamily of nonclustered protocadherin genes, in ASD. PCDH10 expression is enriched in the basolateral amygdala, a brain region implicated in the social deficits of ASD. Previous reports indicate that Pcdh10 plays a role in axon outgrowth and glutamatergic synapse elimination, but its roles in social behaviors and amygdala neuronal connectivity are unknown. We hypothesized that haploinsufficiency of Pcdh10 would reduce social approach behavior and alter the structure and function of amygdala circuits. METHODS: Mice lacking one copy of Pcdh10 (Pcdh10+/-) and wild-type littermates were assessed for social approach and other behaviors. The lateral/basolateral amygdala was assessed for dendritic spine number and morphology, and amygdala circuit function was studied using voltage-sensitive dye imaging. Expression of Pcdh10 and N-methyl-D-aspartate receptor (NMDAR) subunits was assessed in postsynaptic density fractions of the amygdala. RESULTS: Male Pcdh10+/- mice have reduced social approach behavior, as well as impaired gamma synchronization, abnormal spine morphology, and reduced levels of NMDAR subunits in the amygdala. Social approach deficits in Pcdh10+/- male mice were rescued with acute treatment with the NMDAR partial agonist d-cycloserine. CONCLUSIONS: Our studies reveal that male Pcdh10+/- mice have synaptic and behavioral deficits, and establish Pcdh10+/- mice as a novel genetic model for investigating neural circuitry and behavioral changes relevant to ASD.


Subject(s)
Amygdala/physiopathology , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/physiopathology , Cadherins/physiology , Social Behavior , Amygdala/metabolism , Amygdala/pathology , Animals , Autism Spectrum Disorder/psychology , Behavior, Animal/physiology , Cadherins/genetics , Dendritic Spines/pathology , Disease Models, Animal , Electric Stimulation , Female , Gamma Rhythm , Haploinsufficiency , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , Post-Synaptic Density/metabolism , Protocadherins , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Vocalization, Animal
3.
Mol Brain ; 9: 38, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27080392

ABSTRACT

BACKGROUND: Activation of NMDA receptors can induce iron movement into neurons by the small GTPase Dexras1 via the divalent metal transporter 1 (DMT1). This pathway under pathological conditions such as NMDA excitotoxicity contributes to metal-catalyzed reactive oxygen species (ROS) generation and neuronal cell death, and yet its physiological role is not well understood. RESULTS: We found that genetic and pharmacological ablation of this neuronal iron pathway in the mice increased glutamatergic transmission. Voltage sensitive dye imaging of hippocampal slices and whole-cell patch clamping of synaptic currents, indicated that the increase in excitability was due to synaptic modification of NMDA receptor activity via modulation of the PKC/Src/NR2A pathway. Moreover, we identified that lysosomal iron serves as a main source for intracellular iron signaling modulating glutamatergic excitability. CONCLUSIONS: Our data indicates that intracellular iron is dynamically regulated in the neurons and robustly modulate synaptic excitability under physiological condition. Since NMDA receptors play a central role in synaptic neurophysiology, plasticity, neuronal homeostasis, neurodevelopment as well as in the neurobiology of many diseases, endogenous iron is therefore likely to have functional relevance to each of these areas.


Subject(s)
Iron/metabolism , Lysosomes/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , ras Proteins/metabolism , Animals , Cytosol/drug effects , Cytosol/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hydrazines , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Iron Chelating Agents/pharmacology , Lysosomes/drug effects , Mice, Inbred C57BL , Models, Biological , Phosphorylation/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , src-Family Kinases/metabolism
4.
Yale J Biol Med ; 88(1): 25-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25745372

ABSTRACT

Autism spectrum disorders (ASD) are characterized by social impairments and restricted/stereotyped behaviors and currently affect an estimated 1 in 68 children aged 8 years old. While there has been substantial recent focus on ASD in research, both the biological pathology and, perhaps consequently, a fully effective treatment have yet to be realized. What has remained throughout is the hypothesis that ASD has neurobiological underpinnings and the observation that both the phenotypic expression and likely the underlying etiology is highly heterogeneous. Given the neurodevelopmental basis of ASD, a biologically based marker (biomarker) could prove useful not only for diagnostic and prognostic purposes, but also for stratification and response indices for pharmaceutical development. In this review, we examine the current state of the field for MEG-related biomarkers in ASD. We describe several potential biomarkers (middle latency delays [M50/M100], mismatch negativity latency, gamma-band oscillatory activity), and investigate their relation to symptomology, core domains of dysfunction (e.g., language impairment), and putative biological underpinnings.


Subject(s)
Autism Spectrum Disorder/physiopathology , Biomarkers/metabolism , Electrophysiological Phenomena , Magnetoencephalography , Animals , Disease Models, Animal , Humans , Language
5.
Biol Psychiatry ; 77(6): 556-68, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25156700

ABSTRACT

BACKGROUND: Neuronal activity at gamma frequency is impaired in schizophrenia (SZ) and is considered critical for cognitive performance. Such impairments are thought to be due to reduced N-methyl-D-aspartate receptor (NMDAR)-mediated inhibition from parvalbumin interneurons, rather than a direct role of impaired NMDAR signaling on pyramidal neurons. However, recent studies suggest a direct role of pyramidal neurons in regulating gamma oscillations. In particular, a computational model has been proposed in which phasic currents from pyramidal cells could drive synchronized feedback inhibition from interneurons. As such, impairments in pyramidal neuron activity could lead to abnormal gamma oscillations. However, this computational model has not been tested experimentally and the molecular mechanisms underlying pyramidal neuron dysfunction in SZ remain unclear. METHODS: In the present study, we tested the hypothesis that SZ-related phenotypes could arise from reduced NMDAR signaling in pyramidal neurons using forebrain pyramidal neuron specific NMDA receptor 1 knockout mice. RESULTS: The mice displayed increased baseline gamma power, as well as sociocognitive impairments. These phenotypes were associated with increased pyramidal cell excitability due to changes in inherent membrane properties. Interestingly, mutant mice showed decreased expression of GIRK2 channels, which has been linked to increased neuronal excitability. CONCLUSIONS: Our data demonstrate for the first time that NMDAR hypofunction in pyramidal cells is sufficient to cause electrophysiological, molecular, neuropathological, and behavioral changes related to SZ.


Subject(s)
Brain/physiology , Nerve Tissue Proteins/metabolism , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Beta Rhythm/physiology , Cholecystokinin/metabolism , Evoked Potentials, Auditory , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gamma Rhythm/physiology , Glutamate Decarboxylase/metabolism , Memory, Short-Term/physiology , Mice, Knockout , Nerve Tissue Proteins/genetics , Nesting Behavior/physiology , Neural Pathways/physiology , Parvalbumins/metabolism , Prosencephalon/physiology , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Social Behavior , Somatostatin/metabolism , Spatial Memory/physiology , Theta Rhythm/physiology
6.
Brain Struct Funct ; 220(1): 419-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24218106

ABSTRACT

Rett Syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl-CpG-binding protein 2 (MECP2) gene. Affected individuals develop motor deficits including stereotypic hand movements, impaired motor learning and difficulties with movement. To understand the neural mechanisms of motor deficits in RTT, we characterized the molecular and cellular phenotypes in the striatum, the major input nucleus of the basal ganglia that controls psychomotor function, in mice carrying a null allele of Mecp2. These mice showed significant hypoactivity associated with impaired motor coordination and motor skill learning. We found that dopamine content was significantly reduced in the striatum of Mecp2 null mice. Reduced dopamine was accompanied by down-regulation of tyrosine hydroxylase and up-regulation of dopamine D2 receptors, particularly in the rostral striatum. We also observed that loss of MeCP2 induced compartment-specific alterations in the striatum, including reduced expression of µ-opioid receptors in the striosomes and increased number of calbindin-positive neurons in the striatal matrix. The total number of parvalbumin-positive interneurons and their dendritic arborization were also significantly increased in the striatum of Mecp2 null mice. Together, our findings support that MeCP2 regulates a unique set of genes critical for modulating motor output of the striatum, and that aberrant structure and function of the striatum due to MeCP2 deficiency may underlie the motor deficits in RTT.


Subject(s)
Corpus Striatum/pathology , Methyl-CpG-Binding Protein 2/genetics , Psychomotor Disorders/etiology , Psychomotor Disorders/pathology , Rett Syndrome/complications , Rett Syndrome/genetics , Age Factors , Animals , Animals, Newborn , Calbindins/metabolism , Chromatography, High Pressure Liquid , Disease Models, Animal , Dopamine/genetics , Dopamine/metabolism , Exploratory Behavior/physiology , Female , Gene Expression Regulation/genetics , Genotype , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Rotarod Performance Test , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
7.
Front Cell Neurosci ; 8: 414, 2014.
Article in English | MEDLINE | ID: mdl-25538564

ABSTRACT

Most recent estimates indicate that 1 in 68 children are affected by an autism spectrum disorder (ASD). Though decades of research have uncovered much about these disorders, the pathological mechanism remains unknown. Hampering efforts is the seeming inability to integrate findings over the micro to macro scales of study, from changes in molecular, synaptic and cellular function to large-scale brain dysfunction impacting sensory, communicative, motor and cognitive activity. In this review, we describe how studies focusing on neuronal circuit function provide unique context for identifying common neurobiological disease mechanisms of ASD. We discuss how recent EEG and MEG studies in subjects with ASD have repeatedly shown alterations in ensemble population recordings (both in simple evoked related potential latencies and specific frequency subcomponents). Because these disease-associated electrophysiological abnormalities have been recapitulated in rodent models, studying circuit differences in these models may provide access to abnormal circuit function found in ASD. We then identify emerging in vivo and ex vivo techniques, focusing on how these assays can characterize circuit level dysfunction and determine if these abnormalities underlie abnormal clinical electrophysiology. Such circuit level study in animal models may help us understand how diverse genetic and environmental risks can produce a common set of EEG, MEG and anatomical abnormalities found in ASD.

8.
Neurobiol Dis ; 67: 79-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24686304

ABSTRACT

Insulin resistance and other features of the metabolic syndrome are increasingly recognized for their effects on cognitive health. To ascertain mechanisms by which this occurs, we fed mice a very high fat diet (60% kcal by fat) for 17days or a moderate high fat diet (HFD, 45% kcal by fat) for 8weeks and examined changes in brain insulin signaling responses, hippocampal synaptodendritic protein expression, and spatial working memory. Compared to normal control diet mice, cerebral cortex tissues of HFD mice were insulin-resistant as evidenced by failed activation of Akt, S6 and GSK3ß with ex-vivo insulin stimulation. Importantly, we found that expression of brain IPMK, which is necessary for mTOR/Akt signaling, remained decreased in HFD mice upon activation of AMPK. HFD mouse hippocampus exhibited increased expression of serine-phosphorylated insulin receptor substrate 1 (IRS1-pS(616)), a marker of insulin resistance, as well as decreased expression of PSD-95, a scaffolding protein enriched in post-synaptic densities, and synaptopodin, an actin-associated protein enriched in spine apparatuses. Spatial working memory was impaired as assessed by decreased spontaneous alternation in a T-maze. These findings indicate that HFD is associated with telencephalic insulin resistance and deleterious effects on synaptic integrity and cognitive behaviors.


Subject(s)
Brain/metabolism , Dendrites/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance , Spatial Memory/physiology , Synapses/metabolism , Animals , Hyperglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Rats , Signal Transduction
9.
Neuropsychopharmacology ; 39(7): 1603-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24525709

ABSTRACT

NMDA-receptor (NMDAR) hypofunction is strongly implicated in the pathophysiology of schizophrenia. Several convergent lines of evidence suggest that net excitation propagated by impaired NMDAR signaling on GABAergic interneurons may be of particular interest in mediating several aspects of schizophrenia. However, it is unclear which behavioral domains are governed by a net increase of excitation and whether modulating downstream GABAergic signaling can reverse neural and thus behavioral deficits. The current study determines the selective contributions of NMDAR dysfunction on PV-containing interneurons to electrophysiological, cognitive, and negative-symptom-related behavioral phenotypes of schizophrenia using mice with a PVcre-NR1flox-driven ablation of NR1 on PV-containing interneurons. In addition, we assessed the efficacy of one agent that directly modulates GABAergic signaling (baclofen) and one agent that indirectly modifies NMDAR-mediated signaling through antagonism of mGluR5 receptors (2-methyl-6-(phenylethynyl) pyridine (MPEP)). The data indicate that loss of NMDAR function on PV interneurons impairs self-care and sociability while increasing N1 latency and baseline gamma power, and reducing induction and maintenance of long-term potentiation. Baclofen normalized baseline gamma power without corresponding effects on behavior. MPEP further increased N1 latency and reduced social behavior in PVcre/NR1+/+ mice. These two indices were negatively correlated before and following MPEP such that as N1 latency increases, sociability decreases. This finding suggests a predictive role for N1 latency with respect to social function. Although previous data suggest that MPEP may be beneficial for core features of autism spectrum disorders, current data suggest that such effects require intact function of NMDAR on PV interneurons.


Subject(s)
Brain/pathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Parvalbumins/deficiency , Receptors, N-Methyl-D-Aspartate/metabolism , Self Care , Social Behavior Disorders/pathology , Animals , Baclofen/pharmacology , Disease Models, Animal , Evoked Potentials/drug effects , Evoked Potentials/genetics , Excitatory Amino Acid Antagonists/pharmacology , Exploratory Behavior/physiology , GABA Agonists/pharmacology , Interpersonal Relations , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Parvalbumins/genetics , Pyridines/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Rest , Social Behavior Disorders/genetics
10.
Hippocampus ; 24(2): 204-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24446171

ABSTRACT

Genetic variants in DTNBP1 encoding the protein dysbindin-1 have often been associated with schizophrenia and with the cognitive deficits prominent in that disorder. Because impaired function of the hippocampus is thought to play a role in these memory deficits and because NMDAR-dependent synaptic plasticity in this region is a proposed biological substrate for some hippocampal-dependent memory functions in schizophrenia, we hypothesized that reduced dysbindin-1 expression would lead to impairments in NMDAR-dependent synaptic plasticity and in contextual fear conditioning. Acute slices from male mice carrying 0, 1, or 2 null mutant alleles of the Dtnbp1 gene were prepared, and field recordings from the CA1 striatum radiatum were obtained before and after tetanization of Schaffer collaterals of CA3 pyramidal cells. Mice homozygous for the null mutation in Dtnbp1 exhibited significantly reduced NMDAR-dependent synaptic potentiation compared to wild type mice, an effect that could be rescued by bath application of the NMDA receptor coagonist glycine (10 µM). Behavioral testing in adult mice revealed deficits in hippocampal memory processes. Homozygous null mice exhibited lower conditional freezing, without a change in the response to shock itself, indicative of a learning and memory deficit. Taken together, these results indicate that a loss of dysbindin-1 impairs hippocampal plasticity which may, in part, explain the role dysbindin-1 plays in the cognitive impairments of schizophrenia.


Subject(s)
Carrier Proteins/metabolism , Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/genetics , Analysis of Variance , Animals , Biophysics , Carrier Proteins/genetics , Dysbindin , Dystrophin-Associated Proteins , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Freezing Reaction, Cataleptic/physiology , Hippocampus/cytology , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Synapses/physiology
11.
Neuropsychopharmacology ; 38(10): 2035-47, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23624742

ABSTRACT

Smoking is the largest preventable cause of death in the United States. Furthermore, a recent study found that <10% of quit attempts resulted in continuous abstinence for 1 year. With the introduction of pharmacotherapies like Chantix (varenicline), a selective α4ß2 nicotinic partial agonist, successful quit attempts have significantly increased. Therefore, novel subtype-specific nicotinic drugs, such as sazetidine-A, present a rich area for investigation of therapeutic potential in smoking cessation. The present studies examine the anxiety-related behavioral and functional effects of the nicotinic partial agonists varenicline and sazetidine-A during withdrawal from chronic nicotine in mice. Our studies indicate that ventral hippocampal-specific infusions of sazetidine-A, but not varenicline, are efficacious in reducing nicotine withdrawal-related anxiety-like phenotypes in the novelty-induced hypophagia (NIH) paradigm. To further investigate functional differences between these partial agonists, we utilized voltage-sensitive dye imaging (VSDi) in ventral hippocampal slices to determine the effects of sazetidine-A and varenicline in animals chronically treated with saline, nicotine, or undergoing 24 h withdrawal. These studies demonstrate a functional dissociation of varenicline and sazetidine-A on hippocampal network activity, which is directly related to previous drug exposure. Furthermore, the effects of the nicotinic partial agonists in VSDi assays are significantly correlated with their behavioral effects in the NIH test. These findings highlight the importance of drug history in understanding the mechanisms through which nicotinic compounds may be aiding smoking cessation in individuals experiencing withdrawal-associated anxiety.


Subject(s)
Azetidines/pharmacology , Benzazepines/pharmacology , Nicotine/adverse effects , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Quinoxalines/pharmacology , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Azetidines/administration & dosage , Azetidines/therapeutic use , Benzazepines/administration & dosage , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Drug Partial Agonism , Evoked Potentials/drug effects , Evoked Potentials/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Microinjections , Motor Activity/drug effects , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/therapeutic use , Pyridines/administration & dosage , Pyridines/therapeutic use , Quinoxalines/administration & dosage , Receptors, Nicotinic/metabolism , Smoking Cessation/methods , Substance Withdrawal Syndrome/drug therapy , Up-Regulation , Varenicline
12.
Neuropsychopharmacology ; 37(7): 1671-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22414814

ABSTRACT

Dopaminergic and glutamatergic inputs to the nucleus accumbens shell have a central role in reward processing. Non-contingent cocaine administration generates a number of long-term AMPA receptor-dependent changes in synaptic efficacy. However, the synaptic consequences of cocaine self-administration and the potential role of dopamine in these processes remain unclear. Here, we examined the influence of D1 dopamine receptor (D1DR) activation on excitatory synaptic plasticity in the accumbens shell of adult rats following cocaine self-administration. Our results indicated that during the first 2 days following cocaine exposure both pre- and post-synaptic mechanisms contribute to a net decrease in AMPA receptor-mediated signaling. This is reflected by decreased frequency of miniature EPSCs (mEPSCs) attributable to enhanced cannabinoid receptor activity, decreased mEPSC amplitude, and increased paired-pulse ratio of evoked EPSCs. In contrast, the only changes observed in the shell 3-4 weeks following cocaine self-administration were increased mEPSCs amplitudes and AMPA/NMDA ratios. We further found that although these cocaine-induced neuroadaptations during early and late abstinence have different synaptic expression mechanisms, they were normalized by stimulation of D1DRs. Thus, pre-exposure to the D1DR agonist, SKF38393, during the initial period of abstinence increased excitatory synaptic strength, but reduced excitatory signaling after weeks of abstinence. Taken together, these results indicate that the direction of changes in excitatory transmission induced by cocaine self-administration switches over the first few weeks of abstinence. Moreover, D1DRs gate the stability of these cocaine-induced changes at glutamatergic synapses in the accumbens shell by utilizing multiple temporally distinct mechanisms, which has implications for the treatment of cocaine craving and addiction.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Receptors, Dopamine D1/agonists , Synapses/drug effects , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Cocaine/administration & dosage , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Microinjections , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Nucleus Accumbens/physiology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, Dopamine D1/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Self Administration , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
13.
Hippocampus ; 22(2): 230-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21049487

ABSTRACT

Genetic studies have associated deficient function of the serine/threonine kinase Akt1 with schizophrenia. This disorder is associated with developmental, structural, and functional abnormalities of the hippocampus that could be traced to abnormal Akt1 function. To establish a closer connection between Akt1 and hippocampal function, mice with a selective deletion of Akt1 (Akt1(-/-) mice) were examined for physiological and behavioral outcomes dependent on the hippocampus and associated with schizophrenia. Genetic deletion of Akt1 was associated with both impaired proliferative capacity of adult-born hippocampal progenitors and hippocampal long-term potentiation, indicating deficient functions of this brain region associated with neuroplasticity. Moreover, Akt1(-/-) mice demonstrated impairments in contextual fear conditioning and recall of spatial learning, behaviors known to selectively involve the hippocampus. Akt1(-/-) mice also showed reduced prepulse inhibition of the acoustic startle response, a sensorimotor gating response that is perturbed in schizophrenia. Postmortem tissue samples from patients with schizophrenia showed significant reductions of phosphorylated Akt levels in hilar neurons of the dentate gyrus, the neurogenic zone of the hippocampus. Taken together, these results implicate the Akt1 isoform in regulating hippocampal neuroplasticity and cognition and in contributing to the etiology of schizophrenia.


Subject(s)
Hippocampus/metabolism , Learning/physiology , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-akt/metabolism , Schizophrenia/metabolism , Aged , Aged, 80 and over , Animals , Behavior, Animal/physiology , Cell Proliferation , Conditioning, Classical/physiology , Fear , Female , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Reflex, Startle/physiology , Schizophrenia/genetics , Schizophrenia/physiopathology , Spatial Behavior/physiology
14.
Nat Neurosci ; 15(2): 274-83, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22119903

ABSTRACT

Mutations in the MECP2 gene cause the autism spectrum disorder Rett syndrome (RTT). One of the most common MeCP2 mutations associated with RTT occurs at threonine 158, converting it to methionine (T158M) or alanine (T158A). To understand the role of T158 mutations in the pathogenesis of RTT, we generated knockin mice that recapitulate the MeCP2 T158A mutation. We found a causal role for T158A mutation in the development of RTT-like phenotypes, including developmental regression, motor dysfunction, and learning and memory deficits. These phenotypes resemble those present in Mecp2 null mice and manifest through a reduction in MeCP2 binding to methylated DNA and a decrease in MeCP2 protein stability. The age-dependent development of event-related neuronal responses was disrupted by MeCP2 mutation, suggesting that impaired neuronal circuitry underlies the pathogenesis of RTT and that assessment of event-related potentials (ERPs) may serve as a biomarker for RTT and treatment evaluation.


Subject(s)
DNA-Binding Proteins/genetics , Evoked Potentials, Auditory, Brain Stem/genetics , Methyl-CpG-Binding Protein 2/genetics , Mutation/genetics , Threonine/genetics , Acoustic Stimulation/methods , Age Factors , Alanine/genetics , Animals , Cells, Cultured , Cerebral Cortex/cytology , Chromatin Immunoprecipitation , Conditioning, Psychological/physiology , DNA Mutational Analysis , Electroencephalography , Embryo, Mammalian , Exploratory Behavior/physiology , Fear/physiology , Gene Expression Regulation/genetics , Humans , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Neurons/physiology , Spectrum Analysis
15.
Proc Natl Acad Sci U S A ; 108(43): E962-70, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21969553

ABSTRACT

DTNBP1 (dystrobrevin binding protein 1) is a leading candidate susceptibility gene in schizophrenia and is associated with working memory capacity in normal subjects. In schizophrenia, the encoded protein dystrobrevin-binding protein 1 (dysbindin-1) is often reduced in excitatory cortical limbic synapses. We found that reduced dysbindin-1 in mice yielded deficits in auditory-evoked response adaptation, prepulse inhibition of startle, and evoked γ-activity, similar to patterns in schizophrenia. In contrast to the role of dysbindin-1 in glutamatergic transmission, γ-band abnormalities in schizophrenia are most often attributed to disrupted inhibition and reductions in parvalbumin-positive interneuron (PV cell) activity. To determine the mechanism underlying electrophysiological deficits related to reduced dysbindin-1 and the potential role of PV cells, we examined PV cell immunoreactivity and measured changes in net circuit activity using voltage-sensitive dye imaging. The dominant circuit impact of reduced dysbindin-1 was impaired inhibition, and PV cell immunoreactivity was reduced. Thus, this model provides a link between a validated candidate gene and an auditory endophenotypes. Furthermore, these data implicate reduced fast-phasic inhibition as a common underlying mechanism of schizophrenia-associated intermediate phenotypes.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Evoked Potentials, Auditory/physiology , Limbic System/metabolism , Schizophrenia/genetics , Synapses/metabolism , Animals , Dysbindin , Dystrophin-Associated Proteins , Electrophysiology , Evoked Potentials, Auditory/genetics , Female , Genotype , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Parvalbumins
16.
J Physiol ; 589(Pt 8): 1893-903, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21224219

ABSTRACT

Mammalian cortical structures are endowed with the capacity for plasticity, which emerges from a combination of the dynamics of circuit connectivity and function, and the intrinsic function of the neurons within the circuit. However, this capacity is accompanied by a significant risk: the capability to generate seizure discharges is also a property of all mammalian cortices. How do cortical circuits reconcile the requirement to maintain plasticity, but at the same time control seizure initiation? These issues come into particular focus in the hippocampus. The hippocampus is one of the main plasticity engines in the brain, and is also a structure frequently implicated in the generation of epileptic seizures, with temporal lobe epilepsy constituting the most prevalent form of epilepsy in the adult population. One aspect of hippocampal circuitry that is particularly prominent is its intimate interconnections with the entorhinal cortex. These interconnections create a number of excitatory synaptic loops within the limbic system, which, in addition to being important in cognitive function, can support reentrant activation and seizure generation. In the present review, using optical imaging approaches to elucidate circuit processing at high temporal and spatial resolution, we examine how two targets of entorhinal cortical input within the hippocampus, the dentate gyrus and area CA1, regulate these synaptic pathways in ways that can maintain functions important in generation of normal activity patterns, but that dampen the ability of these inputs to generate seizure discharges.


Subject(s)
Epilepsy/physiopathology , Feedback, Physiological , Hippocampus/physiopathology , Molecular Imaging/methods , Neural Pathways/physiopathology , Optics and Photonics , Animals , Brain Waves , Epilepsy/metabolism , Epilepsy/prevention & control , Hippocampus/metabolism , Humans , Interneurons/metabolism , Models, Neurological , Nerve Net/metabolism , Nerve Net/physiopathology , Neural Pathways/metabolism , Neuroglia/metabolism , Neuronal Plasticity , Receptors, GABA-A/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
17.
J Neurosci ; 27(51): 14012-22, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-18094240

ABSTRACT

GABA(A) receptor-mediated inhibition depends on the maintenance of intracellular Cl- concentration ([Cl-]in) at low levels. In neurons in the developing CNS, [Cl-]in is elevated, E(GABA) is depolarizing, and GABA consequently is excitatory. Depolarizing GABAergic synaptic responses may be recapitulated in various neuropathological conditions, including epilepsy. In the present study, rat hippocampal dentate granule cells were recorded using gramicidin perforated patch techniques at varying times (1-60 d) after an epileptogenic injury, pilocarpine-induced status epilepticus (STEP). In normal, non-epileptic animals, these strongly inhibited dentate granule cells act as a gate, regulating hippocampal excitation, controlling seizure initiation and/or propagation. For 2 weeks after STEP, we found that E(GABA) was positively shifted in granule cells. This shift in E(GABA) altered synaptic integration, increased granule cell excitability, and resulted in compromised "gate" function of the dentate gyrus. E(GABA) recovered to control values at longer latencies post-STEP (2-8 weeks), when animals had developed epilepsy. During this period of shifted E(GABA), expression of the Cl- extruding K+/Cl- cotransporter, KCC2 was decreased. Application of the KCC2 blocker, furosemide, to control neurons mimicked E(GABA) shifts evident in granule cells post-STEP. Furthermore, post-STEP and furosemide effects interacted occlusively, both on E(GABA) in granule cells, and on gatekeeper function of the dentate gyrus. This suggests a shared mechanism, reduced KCC2 function. These findings demonstrate that decreased expression of KCC2 persists for weeks after an epileptogenic injury, reducing inhibitory efficacy and enhancing dentate granule cell excitability. This pathophysiological process may constitute a significant mechanism linking injury to the subsequent development of epilepsy.


Subject(s)
Chlorides/physiology , Dentate Gyrus/metabolism , Epilepsy, Temporal Lobe/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Chlorides/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Furosemide/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Symporters/antagonists & inhibitors , Symporters/biosynthesis , Symporters/genetics , Synapses/drug effects , Synapses/physiology , K Cl- Cotransporters
18.
Prog Brain Res ; 163: 235-43, 2007.
Article in English | MEDLINE | ID: mdl-17765722

ABSTRACT

Dentate granule cells are characterized by their low levels of excitability, an important aspect of hippocampal function, which distinguishes them from other principal cells of the hippocampus. This low excitability derives in large part from the degree and nature of GABAergic inhibition evident in the dentate gyrus. Granule cells express a unique and complex assortment of GABA(A) receptor subunits, found in few areas of the brain. Associated with this receptor complexity, granule cells are endowed with both synaptic and extrasynaptic GABA(A) receptors with distinctive properties. In particular, extrasynaptic GABA(A) receptors in granule cells exhibit high affinity for GABA and do not desensitize. This results in activation of a tonic current by ambient levels of GABA present in the extracellular space. This tonic current contributes significantly to the circuit properties of the dentate gyrus. Both synaptic and extrasynaptic GABA(A) receptors exhibit profound dysregulation in animal models of temporal lobe epilepsy, which may contribute to the hippocampal hyperexcitability that defines this disorder.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/physiology , Neural Inhibition/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Humans , Receptors, GABA-A/physiology
19.
J Neurosci ; 26(46): 11850-6, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108158

ABSTRACT

Epilepsy affects 1-2% of the population, with temporal lobe epilepsy (TLE) the most common variant in adults. Clinical and experimental studies have demonstrated hippocampal involvement in the seizures underlying TLE. However, identification of specific functional deficits in hippocampal circuits associated with possible roles in seizure generation remains controversial. Significant attention has focused on anatomic and cellular alterations in the dentate gyrus. The dentate gyrus is a primary gateway regulating cortical input to the hippocampus and, thus, a possible contributor to the aberrant cortical-hippocampal interactions underlying the seizures of TLE. Alternate cortical pathways innervating the hippocampus might also contribute to seizure initiation. Despite this potential importance in TLE, these pathways have received little study. Using simultaneous voltage-sensitive dye imaging and patch-clamp recordings in slices from animals with epilepsy, we assessed the relative degree of synaptic excitation activated by multiple cortical inputs to the hippocampus. Surprisingly, dentate gyrus-mediated regulation of the relay of cortical input to the hippocampus is unchanged in epileptic animals, and input via the Schaffer collaterals is actually decreased despite reduction in Schaffer-evoked inhibition. In contrast, a normally weak direct cortical input to area CA1 of hippocampus, the temporoammonic pathway, exhibits a TLE-associated transformation from a spatially restricted, highly regulated pathway to an excitatory projection with >10-fold increased effectiveness. This dysregulated temporoammonic pathway is critically positioned to mediate generation and/or propagation of seizure activity in the hippocampus.


Subject(s)
Cerebral Cortex/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Neural Inhibition , Neural Pathways/physiopathology , Action Potentials/physiology , Animals , Axons/physiology , Axons/ultrastructure , Cerebral Cortex/cytology , Convulsants/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/physiopathology , Disease Models, Animal , Electric Stimulation , Epilepsy, Temporal Lobe/chemically induced , Excitatory Postsynaptic Potentials/physiology , Fluorescent Dyes , Hippocampus/cytology , Male , Muscarinic Agonists/pharmacology , Neural Inhibition/physiology , Neural Pathways/cytology , Organ Culture Techniques , Patch-Clamp Techniques , Pilocarpine/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
20.
J Neurosci ; 26(33): 8537-48, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-16914680

ABSTRACT

Vesicular GABA and intraterminal glutamate concentrations are in equilibrium, suggesting inhibitory efficacy may depend on glutamate availability. Two main intraterminal glutamate sources are uptake by neuronal glutamate transporters and glutamine synthesized through the astrocytic glutamate-glutamine cycle. We examined the involvement of the glutamate-glutamine cycle in modulating GABAergic synaptic efficacy. In the absence of neuronal activity, disruption of the glutamate-glutamine cycle by blockade of neuronal glutamine transport with alpha-(methylamino) isobutyric acid (MeAIB; 5 mM) or inhibition of glutamine synthesis in astrocytes with methionine sulfoximine (MSO; 1.5 mM) had no effect on miniature IPSCs recorded in hippocampal area CA1 pyramidal neurons. However, after a period of moderate synaptic activity, application of MeAIB, MSO, or dihydrokainate (250 microM; an astrocytic glutamate transporter inhibitor) significantly reduced evoked IPSC (eIPSC) amplitudes. The MSO effect could be reversed by exogenous application of glutamine (5 mM), whereas glutamine could not rescue the eIPSC decreases induced by the neuronal glutamine transporter inhibitor MeAIB. The activity-dependent reduction in eIPSCs by glutamate-glutamine cycle blockers was accompanied by an enhanced blocking effect of the low-affinity GABA(A) receptor antagonist, TPMPA [1,2,5,6-tetrahydropyridin-4-yl)methylphosphinic acid], consistent with diminished GABA release. We further corroborated this hypothesis by examining MeAIB effects on minimal stimulation-evoked quantal IPSCs (meIPSCs). We found that, in MeAIB-containing medium, moderate stimulation induced depression in potency of meIPSCs but no change in release probability, consistent with reduced vesicular GABA content. We conclude that the glutamate-glutamine cycle is a major contributor to synaptic GABA release under physiological conditions, which dynamically regulates inhibitory synaptic strength.


Subject(s)
Glutamates/metabolism , Glutamine/metabolism , Hippocampus/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Amino Acid Transport System X-AG/antagonists & inhibitors , Animals , Astrocytes/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamine/biosynthesis , Glutamine/pharmacology , Hippocampus/cytology , In Vitro Techniques , Male , Methionine Sulfoximine , Neural Inhibition/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Synapses/physiology , beta-Alanine/analogs & derivatives , beta-Alanine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...