Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28451631

ABSTRACT

Fragile X syndrome is a genetic condition resulting from FMR1 gene mutation that leads to intellectual disability, autism-like symptoms, and sensory hypersensitivity. Arbaclofen, a GABA-B agonist, has shown efficacy in some individuals with FXS but has become unavailable after unsuccessful clinical trials, prompting interest in publicly available, racemic baclofen. The present study investigated whether racemic baclofen can remediate abnormalities of neural circuit function, sensory processing, and behavior in Fmr1 knockout mice, a rodent model of fragile X syndrome. Fmr1 knockout mice showed increased baseline and auditory-evoked high-frequency gamma (30-80 Hz) power relative to C57BL/6 controls, as measured by electroencephalography. These deficits were accompanied by decreased T maze spontaneous alternation, decreased social interactions, and increased open field center time, suggestive of diminished working memory, sociability, and anxiety-like behavior, respectively. Abnormal auditory-evoked gamma oscillations, working memory, and anxiety-related behavior were normalized by treatment with baclofen, but impaired sociability was not. Improvements in working memory were evident predominantly in mice whose auditory-evoked gamma oscillations were dampened by baclofen. These findings suggest that racemic baclofen may be useful for targeting sensory and cognitive disturbances in fragile X syndrome.


Subject(s)
Baclofen/pharmacology , Evoked Potentials, Auditory/drug effects , Fragile X Syndrome/complications , GABA-B Receptor Agonists/pharmacology , Mental Disorders/etiology , Mental Disorders/pathology , Acoustic Stimulation , Animals , Disease Models, Animal , Electroencephalography , Evoked Potentials, Auditory/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Interpersonal Relations , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spectrum Analysis
2.
Neurosci Biobehav Rev ; 76(Pt B): 235-253, 2017 05.
Article in English | MEDLINE | ID: mdl-27235081

ABSTRACT

Brains are constantly flooded with sensory information that needs to be filtered at the pre-attentional level and integrated into endogenous activity in order to allow for detection of salient information and an appropriate behavioral response. People with Autism Spectrum Disorder (ASD) or Fragile X Syndrome (FXS) are often over- or under-reactive to stimulation, leading to a wide range of behavioral symptoms. This altered sensitivity may be caused by disrupted sensory processing, signal integration and/or gating, and is often being neglected. Here, we review translational experimental approaches that are used to investigate sensory processing in humans with ASD and FXS, and in relevant rodent models. This includes electroencephalographic measurement of event related potentials, neural oscillations and mismatch negativity, as well as habituation and pre-pulse inhibition of startle. We outline robust evidence of disrupted sensory processing in individuals with ASD and FXS, and in respective animal models, focusing on the auditory sensory domain. Animal models provide an excellent opportunity to examine common mechanisms of sensory pathophysiology in order to develop therapeutics.


Subject(s)
Autism Spectrum Disorder , Fragile X Syndrome , Animals , Brain , Electroencephalography , Humans , Models, Animal
3.
Neuroscience ; 322: 408-15, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26946266

ABSTRACT

Dexras1 is a novel GTPase that acts at a confluence of signaling mechanisms associated with psychiatric and neurological disease including NMDA receptors, NOS1AP and nNOS. Recent work has shown that Dexras1 mediates iron trafficking and NMDA-dependent neurodegeneration but a role for Dexras1 in normal brain function or psychiatric disease has not been studied. To test for such a role, mice with germline knockout (KO) of Dexras1 were assayed for behavioral abnormalities as well as changes in NMDA receptor subunit protein expression. Because Dexras1 is up-regulated during stress or by dexamethasone treatment, we included measures associated with emotion including anxiety and depression. Baseline anxiety-like measures (open field and zero maze) were not altered, nor were depression-like behavior (tail suspension). Measures of memory function yielded mixed results, with no changes in episodic memory (novel object recognition) but a significant decrement on working memory (T-maze). Alternatively, there was an increase in pre-pulse inhibition (PPI), without concomitant changes in either startle amplitude or locomotor activity. PPI data are consistent with the direction of change seen following exposure to dopamine D2 antagonists. An examination of NMDA subunit expression levels revealed an increased expression of the NR2A subunit, contrary to previous studies demonstrating down-regulation of the receptor following antipsychotic exposure (Schmitt et al., 2003) and up-regulation after exposure to isolation rearing (Turnock-Jones et al., 2009). These findings suggest a potential role for Dexras1 in modulating a selective subset of psychiatric symptoms, possibly via its interaction with NMDARs and/or other disease-related binding-partners. Furthermore, data suggest that modulating Dexras1 activity has contrasting effects on emotional, sensory and cognitive domains.


Subject(s)
Anxiety Disorders/metabolism , Memory, Short-Term/physiology , Prepulse Inhibition/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , ras Proteins/metabolism , Animals , Maze Learning/physiology , Memory, Episodic , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Recognition, Psychology/physiology , ras Proteins/genetics
4.
Neuroscience ; 321: 66-76, 2016 May 03.
Article in English | MEDLINE | ID: mdl-26577758

ABSTRACT

Schizophrenia (SCZ) is a disorder characterized by positive symptoms (hallucinations, delusions), negative symptoms (blunted affect, alogia, reduced sociability, and anhedonia), as well as persistent cognitive deficits (memory, concentration, and learning). While the biology underlying subjective experiences is difficult to study, abnormalities in electroencephalographic (EEG) measures offer a means to dissect potential circuit and cellular changes in brain function. EEG is indispensable for studying cerebral information processing due to the introduction of techniques for the decomposition of event-related activity into its frequency components. Specifically, brain activity in the gamma frequency range (30-80Hz) is thought to underlie cognitive function and may be used as an endophenotype to aid in diagnosis and treatment of SCZ. In this review we address evidence indicating that there is increased resting-state gamma power in SCZ. We address how modeling this aspect of the illness in animals may help treatment development as well as providing insights into the etiology of SCZ.


Subject(s)
Disease Models, Animal , Gamma Rhythm , Schizophrenia/physiopathology , Animals , Brain/physiopathology , Humans , Male
5.
Drug Alcohol Depend ; 156: 14-20, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26442907

ABSTRACT

BACKGROUND: Ketamine, an N-methyl-d-aspartate receptor antagonist, is used as a pediatric anesthetic because of its favorable safety profile. It is also being investigated as an antidepressant. Unfortunately, ketamine causes adverse reactions including hallucinations and is associated with a high prevalence of abuse among adolescents. Although chronic ketamine use has been shown to produce cognitive impairments even years following cessation, little is known about its long-term consequences on adolescents. The beta-lactam ceftriaxone has been shown to attenuate alcohol withdrawal, and alleviate early brain injury and memory impairments following subarachnoid hemorrhage. However, its ability to reverse the effects of adolescent ketamine exposure is not known. Previous data indicate that ketamine causes a reduction in the number of Excitatory Amino Acid Transporter Type 2 (EAAT2)-containing astrocytes. Additionally, the beta lactam antibiotic ceftriaxone increased expression of EAAT2. As EAAT2 is a principal mechanism of glutamate clearance from the synapse, the current study tests the hypothesis that ceftriaxone may reverse functional consequences of ketamine exposure. METHODS: We examined the effects of chronic ketamine in juvenile mice as well as reversal by ceftriaxone using electroencephalography (EEG). Subsequently, we assessed the effects of these treatments on markers of astrocyte proliferation, using Glial Fibrillary Acidic Protein (GFAP), and function, as evidenced by EAAT2. RESULTS: Juvenile mice exposed to chronic ketamine showed lasting alterations in EEG measurements as well as markers of astrocyte number and function. These alterations were reversed by ceftriaxone. CONCLUSIONS: Data suggest that ceftriaxone may be able to ameliorate ketamine-induced long-term disturbances in adolescent brains.


Subject(s)
Anesthetics, Dissociative/pharmacology , Anti-Bacterial Agents/pharmacology , Astrocytes/drug effects , Ceftriaxone/pharmacology , Electroencephalography/drug effects , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Animals , Brain/cytology , Brain/drug effects , Cell Count , Electrodes, Implanted , Evoked Potentials/drug effects , Excitatory Amino Acid Transporter 2/genetics , Gamma Rhythm/drug effects , Glial Fibrillary Acidic Protein/genetics , Male , Mice , Mice, Inbred C57BL
6.
Transl Psychiatry ; 5: e635, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26371763

ABSTRACT

Frequent ketamine abuse in adulthood correlates with increased risk of psychosis, as well as cognitive deficits, including disruption of higher-order executive function and memory formation. Although the primary abusers of ketamine are adolescents and young adults, few studies have evaluated its effects on juvenile cognition. Therefore, the current study analyzes the effect of adolescent ketamine exposure on cognitive development. Juvenile mice (4 weeks of age) were exposed to chronic ketamine (20 mg kg(-1), i.p. daily) for 14 days. Mice were tested immediately after exposure in the juvenile period (7 weeks of age) and again as adults (12 weeks of age). Measures included electroencephalography (EEG) in response to auditory stimulation, the social choice test, and a 6-arm radial water maze task. Outcome measures include low-frequency EEG responses, event-related potential (ERP) amplitudes, indices of social behavior and indices of spatial working memory. Juvenile exposure to ketamine was associated with electrophysiological abnormalities in adulthood, particularly in induced theta power and the P80 ERP. The social choice test revealed that ketamine-exposed mice failed to exhibit the same age-related decrease in social interaction time as controls. Ketamine-exposed mice outperformed control mice as juveniles on the radial water maze task, but did not show the same age-related improvement as adult controls. These data support the hypothesis that juvenile exposure to ketamine produces long-lasting changes in brain function that are characterized by a failure to progress along normal developmental trajectories.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Brain/physiopathology , Cognition Disorders/chemically induced , Cognition Disorders/physiopathology , Ketamine/pharmacology , Animals , Cognition/drug effects , Disease Models, Animal , Electroencephalography/drug effects , Electroencephalography/statistics & numerical data , Evoked Potentials, Auditory/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Male , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Reaction Time/drug effects , Time Factors
7.
Mol Psychiatry ; 20(9): 1091-100, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25330739

ABSTRACT

Numerous investigations support decreased glutamatergic signaling as a pathogenic mechanism of schizophrenia, yet the molecular underpinnings for such dysregulation are largely unknown. In the post-mortem dorsolateral prefrontal cortex (DLPFC), we found striking decreases in tyrosine phosphorylation of N-methyl-D aspartate (NMDA) receptor subunit 2 (GluN2) that is critical for neuroplasticity. The decreased GluN2 activity in schizophrenia may not be because of downregulation of NMDA receptors as MK-801 binding and NMDA receptor complexes in postsynaptic density (PSD) were in fact increased in schizophrenia cases. At the postreceptor level, however, we found striking reductions in the protein kinase C, Pyk 2 and Src kinase activity that in tandem can decrease GluN2 activation. Given that Src serves as a hub of various signaling mechanisms affecting GluN2 phosphorylation, we postulated that Src hypoactivity may result from convergent alterations of various schizophrenia susceptibility pathways and thus mediate their effects on NMDA receptor signaling. Indeed, the DLPFC of schizophrenia cases exhibit increased PSD-95 and erbB4 and decreased receptor-type tyrosine-protein phosphatase-α (RPTPα) and dysbindin-1, each of which reduces Src activity via protein interaction with Src. To test genomic underpinnings for Src hypoactivity, we examined genome-wide association study results, incorporating 13 394 cases and 34 676 controls. We found no significant association of individual variants of Src and its direct regulators with schizophrenia. However, a protein-protein interaction-based network centered on Src showed significant enrichment of gene-level associations with schizophrenia compared with other psychiatric illnesses. Our results together demonstrate striking decreases in NMDA receptor signaling at the postreceptor level and propose Src as a nodal point of convergent dysregulations affecting NMDA receptor pathway via protein-protein associations.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , src-Family Kinases/metabolism , Animals , Brain/metabolism , Brain/pathology , Case-Control Studies , Gene Expression Regulation , Genome-Wide Association Study , Humans , Mice , Mice, Knockout , Neuronal Plasticity , Phosphorylation , Post-Synaptic Density/genetics , Post-Synaptic Density/metabolism , Prefrontal Cortex/metabolism , Protein Interaction Maps , Schizophrenia/enzymology , Schizophrenia/pathology , Signal Transduction , src-Family Kinases/genetics
8.
Drug Alcohol Depend ; 134: 123-127, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24210161

ABSTRACT

BACKGROUND: Increased susceptibility to cognitive impairment or psychosis in adulthood is associated with adolescent drug abuse. Studies in adults have identified impairments in attention and memory, and changes in EEG, as common consequences of ketamine abuse. In contrast, the effects of ketamine on the juvenile brain have not been extensively tested. This is a significant omission, since abuse of ketamine is often observed within this age group. OBJECTIVES: Juvenile mice (4-6 weeks of age) were administered ketamine (20mg/kg) for 14 days. EEG was assessed in response to auditory stimulation both at one week following ketamine exposure at 7 weeks of age (juvenile) and again at 12 weeks of age (adult). EEG was analyzed for baseline activity, event-related power and event-related potentials (ERPs). RESULTS: While no effects of ketamine exposure were observed during the juvenile period, significant reductions in amplitude of the P20 ERP component and event-related gamma power were seen following ketamine when re-tested as adults. In contrast, reductions in event-related theta were seen in ketamine-exposed mice at both time points. CONCLUSIONS: Age related deficits in electrophysiological components such as P20 or event-related gamma may be due to an interruption of normal neural maturation. Reduction of NMDAR signaling during adolescence leads to delayed-onset disruption of gamma oscillations and the P20 component of the ERP. Further, delayed onset of impairment following adolescent ketamine abuse suggests that methods could be developed to detect and treat the early effects of drug exposure prior to the onset of disability.


Subject(s)
Electroencephalography/drug effects , Ketamine/toxicity , Reaction Time/drug effects , Acoustic Stimulation/methods , Age Factors , Animals , Electrodes, Implanted , Electroencephalography/methods , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Ketamine/administration & dosage , Male , Mice , Mice, Inbred C3H , Reaction Time/physiology
9.
Neuroimage ; 77: 93-104, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23558094

ABSTRACT

Pavlovian fear conditioning has been thoroughly studied in the visual, auditory and somatosensory domain, but evidence is scarce with regard to the chemosensory modality. Under the assumption that Pavlovian conditioning relies on the supra-modal mechanism of salience attribution, the present study was set out to attest the existence of chemosensory aversive conditioning in humans as a specific instance of salience attribution. fMRI was performed in 29 healthy subjects during a differential aversive conditioning paradigm. Two odors (rose, vanillin) served as conditioned stimuli (CS), one of which (CS+) was intermittently coupled with intranasally administered CO2. On the neural level, a robust differential response to the CS+ emerged in frontal, temporal, occipito-parietal and subcortical brain regions, including the amygdala. These changes were paralleled by the development of a CS+-specific connectivity profile of the anterior midcingulate cortex (aMCC), which is a key structure for processing salience information in order to guide adaptive response selection. Increased coupling could be found between key nodes of the salience network (anterior insula, neo-cerebellum) and sensorimotor areas, representing putative input and output structures of the aMCC for exerting adaptive motor control. In contrast, behavioral and skin conductance responses did not show significant effects of conditioning, which has been attributed to contingency unawareness. These findings imply substantial similarities of conditioning involving chemosensory and other sensory modalities, and suggest that salience attribution and adaptive control represent a general, modality-independent principle underlying Pavlovian conditioning.


Subject(s)
Brain Mapping , Brain/physiology , Conditioning, Classical/physiology , Olfactory Perception/physiology , Adult , Female , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging , Male , Pain/physiopathology , Trigeminal Nerve/physiology
10.
Transl Psychiatry ; 2: e142, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22806213

ABSTRACT

Reduced N-methyl-D-aspartate-receptor (NMDAR) signaling has been associated with schizophrenia, autism and intellectual disability. NMDAR-hypofunction is thought to contribute to social, cognitive and gamma (30-80 Hz) oscillatory abnormalities, phenotypes common to these disorders. However, circuit-level mechanisms underlying such deficits remain unclear. This study investigated the relationship between gamma synchrony, excitatory-inhibitory (E/I) signaling, and behavioral phenotypes in NMDA-NR1(neo-/-) mice, which have constitutively reduced expression of the obligate NR1 subunit to model disrupted developmental NMDAR function. Constitutive NMDAR-hypofunction caused a loss of E/I balance, with an increase in intrinsic pyramidal cell excitability and a selective disruption of parvalbumin-expressing interneurons. Disrupted E/I coupling was associated with deficits in auditory-evoked gamma signal-to-noise ratio (SNR). Gamma-band abnormalities predicted deficits in spatial working memory and social preference, linking cellular changes in E/I signaling to target behaviors. The GABA(B)-receptor agonist baclofen improved E/I balance, gamma-SNR and broadly reversed behavioral deficits. These data demonstrate a clinically relevant, highly translatable neural-activity-based biomarker for preclinical screening and therapeutic development across a broad range of disorders that share common endophenotypes and disrupted NMDA-receptor signaling.


Subject(s)
Baclofen/pharmacology , Evoked Potentials, Auditory/genetics , Exploratory Behavior/physiology , GABA-B Receptor Agonists/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Social Behavior , Synaptic Potentials/genetics , Animals , Autistic Disorder/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Exploratory Behavior/drug effects , Fluorobenzenes/pharmacology , In Situ Hybridization , Intellectual Disability/physiopathology , Interneurons/metabolism , Interneurons/pathology , Male , Mice , Mice, Transgenic , Parvalbumins/metabolism , Phenotype , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Risperidone/pharmacology , Schizophrenia/physiopathology , Synaptic Potentials/drug effects , Synaptic Potentials/physiology , Triazoles/pharmacology
11.
Genes Brain Behav ; 11(6): 740-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22726567

ABSTRACT

Reduced NMDA-receptor (NMDAR) function has been implicated in the pathophysiology of neuropsychiatric disease, most strongly in schizophrenia but also recently in autism spectrum disorders (ASD). To determine the direct contribution of NMDAR dysfunction to disease phenotypes, a mouse model with constitutively reduced expression of the obligatory NR1 subunit has been developed and extensively investigated. Adult NR1(neo-/-) mice show multiple abnormal behaviors, including reduced social interactions, locomotor hyperactivity, self-injury, deficits in prepulse inhibition (PPI) and sensory hypersensitivity, among others. Whereas such phenotypes have largely been interpreted in the context of schizophrenia, these behavioral abnormalities are rather non-specific and are frequently present across models of diseases characterized by negative symptom domains. This study investigated auditory electrophysiological and behavioral paradigms relevant to autism, to determine whether NMDAR hypofunction may be more consistent with adult ASD-like phenotypes. Indeed, transgenic mice showed behavioral deficits relevant to all core ASD symptoms, including decreased social interactions, altered ultrasonic vocalizations and increased repetitive behaviors. NMDAR disruption recapitulated clinical endophenotypes including reduced PPI, auditory-evoked response N1 latency delay and reduced gamma synchrony. Auditory electrophysiological abnormalities more closely resembled those seen in clinical studies of autism than schizophrenia. These results suggest that NMDAR hypofunction may be associated with a continuum of neuropsychiatric diseases, including schizophrenia and autism. Neural synchrony abnormalities suggest an imbalance of glutamatergic and GABAergic coupling and may provide a target, along with behavioral phenotypes, for preclinical screening of novel therapeutics.


Subject(s)
Autistic Disorder/genetics , Behavior, Animal/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Social Behavior , Animals , Autistic Disorder/epidemiology , Disease Models, Animal , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Schizophrenia/epidemiology
12.
Genes Brain Behav ; 8(7): 661-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563516

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play a pivotal role in excitatory neurotransmission, synaptic plasticity and brain development. Clinical and experimental evidence suggests a dysregulation of NMDAR function and glutamatergic pathways in the pathophysiology of schizophrenia. We evaluated electrophysiological and behavioral properties of NMDAR deficiency utilizing mice that express only 5-10% of the normal level of NMDAR NR1 subunit. Auditory and visual event related potentials yielded significantly increased amplitudes for the P20 and N40 components in NMDAR deficient (NR1(neo)-/-) mice suggesting decreased inhibitory tone. Compared to wild types, NR1(neo)-/- mice spent less time in social interactions and showed reduced nest building. NR1(neo)-/- mice displayed a preference for open arms of a zero maze and central zone of an open field, possibly reflecting decreased anxiety-related behavioral inhibition. However, locomotor activity did not differ between groups in either home cage environment or during behavioral testing. NR1(neo)-/- mice displayed hyperactivity only when placed in a large unfamiliar environment, suggesting that neither increased anxiety nor non-specific motor activation accounts for differential behavioral patterns. Data suggest that NMDAR NR1 deficiency causes disinhibition in sensory processing as well as reduced behavioral inhibition and impaired social interactions. The behavioral signature in NR1(neo)-/- mice supports the impact of impaired NMDAR function in a mouse model with possible relevance to negative symptoms in schizophrenia.


Subject(s)
Brain Chemistry/genetics , Brain/metabolism , Genetic Predisposition to Disease/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Schizophrenia/metabolism , Animals , Anxiety/genetics , Auditory Diseases, Central/genetics , Auditory Diseases, Central/metabolism , Auditory Diseases, Central/physiopathology , Behavior, Animal/physiology , Brain/physiopathology , Disease Models, Animal , Evoked Potentials/genetics , Female , Genotype , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neural Inhibition/genetics , Perceptual Disorders/genetics , Perceptual Disorders/metabolism , Perceptual Disorders/physiopathology , Phenotype , Schizophrenia/physiopathology , Social Behavior , Visual Pathways/metabolism , Visual Pathways/physiopathology
13.
Neuroscience ; 158(2): 705-12, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19015010

ABSTRACT

INTRODUCTION: Electrophysiological responses to auditory stimuli have provided a useful means of elucidating mechanisms and evaluating treatments in psychiatric disorders. Deficits in gating during paired-click tasks and lack of mismatch negativity following deviant stimuli have been well characterized in patients with schizophrenia. Recently, analyses of basal, induced, and evoked frequency oscillations have gained support as additional measures of cognitive processing in patients and animal models. The purpose of this study is to examine frequency oscillations in mice across the theta (4-7.5 Hz) and gamma (31-61 Hz) bands in the context of N-methyl-d-aspartic acid receptor (NMDAR) hypofunction and dopaminergic hyperactivity, both of which are thought to serve as pharmacological models of schizophrenia. EXPERIMENTAL PROCEDURES: Electroencephalograms (EEG) were recorded from mice in five treatment groups that consisted of haloperidol, risperidone, amphetamine, ketamine, or ketamine plus haloperidol during an auditory task. Basal, induced and evoked powers in both frequencies were calculated. RESULTS: Ketamine increased basal power in the gamma band and decreased the evoked power in the theta band. The increase in basal gamma was not blocked by treatment with a conventional antipsychotic. No other treatment group was able to fully reproduce this pattern in the mice. CONCLUSIONS: Ketamine-induced alterations in EEG power spectra are consistent with abnormalities in the theta and gamma frequency ranges reported in patients with schizophrenia. Our findings support the hypothesis that NMDAR hypofunction contributes to the deficits in schizophrenia and that the dopaminergic pathways alone may not account for these changes.


Subject(s)
Biological Clocks/drug effects , Electroencephalography , Evoked Potentials, Auditory/drug effects , Excitatory Amino Acid Antagonists/adverse effects , Ketamine/adverse effects , Schizophrenia/chemically induced , Acoustic Stimulation/methods , Amphetamine/administration & dosage , Animals , Antipsychotic Agents/pharmacology , Central Nervous System Stimulants/administration & dosage , Disease Models, Animal , Drug Interactions , Haloperidol/pharmacology , Male , Mice , Mice, Inbred C57BL , Reaction Time/drug effects , Risperidone/pharmacology , Schizophrenia/drug therapy , Schizophrenia/physiopathology
14.
Neuroscience ; 157(1): 95-104, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18835334

ABSTRACT

PURPOSE: Chemotherapeutic agents are known to produce persistent cognitive deficits in cancer patients. However, little progress has been made in developing animal models to explore underlying mechanisms and potential therapeutic interventions. We report an electrophysiological model of chemotherapy-induced cognitive deficits using a sensory gating paradigm, to correspond with performance in two behavioral tasks. EXPERIMENTAL DESIGN: Mice received four weekly injections of methotrexate and 5-fluorouracil. Whole-brain event-related potentials (ERPs) were recorded throughout using a paired-click paradigm. Mice underwent contextual fear conditioning (CFC) and novel-object recognition testing (NOR). RESULTS: Chemotherapy-treated animals showed significantly impaired gating 5 weeks after drug treatments began, as measured by the ratio of the first positive peak in the ERP (P1) minus the first negative peak (N1) between first and second auditory stimuli. There was no effect of drug on the amplitude of P1-N1 or latency of P1. The drug-treated animals also showed significantly increased freezing during fear conditioning and increased exploration without memory impairment during novel object recognition. CONCLUSIONS: Chemotherapy causes decreased ability to gate incoming auditory stimuli, which may underlie associated cognitive impairments. These gating deficits were associated with a hyperactive response to fear conditioning and reduced adaptation to novel objects, suggesting an additional component of emotional dysregulation. However, amplitudes and latencies of ERP components were unaffected, as was NOR performance, highlighting the subtle nature of these deficits.


Subject(s)
Antineoplastic Agents/toxicity , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Animals , Antimetabolites, Antineoplastic/toxicity , Auditory Perception/drug effects , Cognition Disorders/chemically induced , Electroencephalography , Electrophysiology , Evoked Potentials/physiology , Evoked Potentials, Auditory/drug effects , Fear/drug effects , Fluorouracil/toxicity , Male , Methotrexate/toxicity , Mice , Mice, Inbred C57BL , Neurotoxicity Syndromes/psychology , Recognition, Psychology/drug effects
15.
J Pharmacol Exp Ther ; 326(1): 230-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18420599

ABSTRACT

Antipsychotic medications function through antagonism of D2 dopamine receptors. Blockade of D2 receptors causes an increase in intracellular cAMP, a ubiquitous second messenger. Inhibition of phosphodiesterase (PDE) activity, a family of enzymes that degrade cyclic nucleotides, causes the same effect. The conceptual linkage between dopamine D2 receptors and PDE activity via cAMP suggests a possible therapeutic potential for PDE inhibitors in schizophrenia. The limited number of studies in support of this hypothesis used rolipram, a specific inhibitor of the PDE4 family. In this study, we investigated the impact of 4-(3-butoxy-4-methoxybenzyl)-2-imidazolidinone (RO-20-1724), another PDE4-specific inhibitor, on auditory event-related potentials (ERPs), prepulse inhibition (PPI) of the startle reflex, and locomotor activity in mice. The ability to reverse amphetamine-induced alterations in ERPs and PPI was used as a model for psychosis. ERPs after RO-20-1724 revealed increased amplitude for the P20 and N40 ERP components. RO-20-1724 reversed the disruptive effect of amphetamines on ERPs and restored gating at a dose that did not impair locomotor activity. However, RO-20-1724 failed to reverse a amphetamine-induced decrease of PPI. Inconsistent results between these two psychosis models suggest that pure sensory processing, as measured with auditory ERPs, may be more sensitive to the effects of intracellular cAMP than sensorimotor effects as assessed with PPI. It remains unclear whether antipsychotic-like properties are a common feature of PDE4 inhibition, or if they are restricted to the pharmacological profile of rolipram. Future studies should examine how PDE4 subtype specificity might contribute to differences between rolipram and RO-20-1724 in sensorimotor gating.


Subject(s)
4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Antipsychotic Agents/pharmacology , Evoked Potentials, Auditory/drug effects , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Reflex, Startle/drug effects , Acoustic Stimulation/methods , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Drug Evaluation, Preclinical/methods , Evoked Potentials, Auditory/physiology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Reflex, Startle/physiology
16.
Schizophr Res ; 99(1-3): 312-23, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18155880

ABSTRACT

Verbal fluency deficits in schizophrenia are difficult to interpret because the tasks are multi-factorial and groups differ in total words generated. We manipulated retrieval and switching demands by requiring alternation between over-learned sequences in which retrieval is relatively automatic (OS) and semantic categories requiring increased retrieval effort (SC). Controlled processing was also manipulated by including switching and non-switching conditions, and formal thought disorder (FTD) was assessed with the communication disorders index (CDI). The OS/SC semantic fluency paradigm was administered during fMRI to 13 patients with schizophrenia and 14 matched controls. Images were acquired on a 3 Tesla Siemens scanner using compressed image acquisition to allow for cued overt word production. Subjects alternated between OS, SC, OS-switch, SC-switch, and baseline blocks. Images were pre-processed in SPM-2, and a two-stage random effects analysis tested within and between group contrasts. There were no group performance differences. fMRI analysis did not reveal any group differences during the OS non-switching condition. Both groups produced expected activation in bilateral prefrontal and inferior parietal regions. However, during the SC condition patients had greater activation than controls in left prefrontal, right anterior cingulate, right superior temporal, bilateral thalamus, and left parietal regions. There was also evidence of patient over-activation in prefrontal, superior temporal, superior parietal, and visual association areas when a switching component was added. FTD was negatively correlated with BOLD response in the right anterior cingulate, cuneus and superior frontal gyrus during increased retrieval demand, and positively correlated with fMRI activation in the left lingual gyrus, right fusiform gyrus and left superior parietal lobule during increased switching demand. These results indicate that patients are able to successfully perform effortful semantic fluency tasks during non-speeded conditions. When retrieval is relatively automatic there does not appear to be an effect of schizophrenia on fMRI response. However, when retrieval and controlled processing demands increase, patients have greater activation than controls despite unimpaired task performance. This inefficient BOLD response may explain why patients are slower and less accurate on standard self-paced fluency tasks.


Subject(s)
Arousal/physiology , Attention/physiology , Brain/physiopathology , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Mental Recall/physiology , Schizophrenia/physiopathology , Schizophrenic Psychology , Semantics , Verbal Behavior/physiology , Adult , Brain Mapping , Cerebral Cortex/physiopathology , Cognition Disorders/diagnosis , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Dominance, Cerebral/physiology , Female , Humans , Male , Nerve Net/physiopathology , Schizophrenia/diagnosis , Speech Production Measurement , Thinking/physiology
17.
Schizophr Res ; 98(1-3): 66-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17765477

ABSTRACT

BACKGROUND: Non-adherence with medication is a critical limitation in current long-term treatment of schizophrenia and a primary factor in poor quality-of-life outcomes. However, few treatments have addressed this shortcoming using an implantable drug delivery approach. The goal of this study was to provide in vitro and in vivo proof of concept for a long-term implantable risperidone delivery system in mice. METHODS: Implantable formulations of risperidone were created using the biodegradable polymer Poly Lactic co Glycolic Acid (PLGA) combined with various drug loads. Implant bioactivity was tested using in vitro release and stability studies, as well as in vivo pharmacokinetic and behavioral studies in mice. RESULTS: The pattern of risperidone release is influenced by various parameters, including polymer composition and drug load. In vitro measures demonstrate that risperidone is stable in implants under physiological conditions. Behavioral measures demonstrate the bioactivity of risperidone implants delivering 3 mg/kg/day in mice, while pharmacokinetic analyses indicate that reversibility is maintained throughout the delivery interval. CONCLUSIONS: The current report suggests that implantable formulations are a viable approach to providing long-term delivery of antipsychotic medications based on in vivo animal studies and pharmacokinetics. Implantable medications demonstrated here can last two months or longer while maintaining coherence and removability past full release, suggesting a potential paradigm shift in the long-term treatment of schizophrenia.


Subject(s)
Drug Delivery Systems/methods , Drug Implants/pharmacokinetics , Models, Animal , Risperidone/pharmacokinetics , Animals , Behavior, Animal/drug effects , Biocompatible Materials , Disease Models, Animal , Drug Delivery Systems/instrumentation , Drug Design , Drug Implants/pharmacology , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Humans , In Vitro Techniques , Lactic Acid , Mice , Mice, Inbred C57BL , Patient Compliance , Polyesters , Polyglycolic Acid , Polymers , Reflex, Startle/drug effects , Reflex, Startle/physiology , Risperidone/pharmacology , Schizophrenia/drug therapy
18.
Neuroscience ; 144(1): 239-46, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17081698

ABSTRACT

Currently available antipsychotic medications work primarily by antagonizing D2 dopamine receptors, thus raising intracellular cAMP levels. We hypothesized that intracellular stimulation of cAMP levels in the CNS would have similar effects to treatment with antipsychotic medication. To test this hypothesis, we studied the effect of an acute treatment of rolipram, an inhibitor of type 4 phosphodiesterases that degrade cAMP, on acoustic startle and prepulse inhibition (PPI) of the acoustic startle response in C57BL/6J mice known to exhibit poor PPI. PPI is disrupted in schizophrenia patients, and the ability of a drug to increase PPI in mice is predictive of antipsychotic efficacy. We show here that acute treatment with rolipram significantly increases PPI at doses that do not alter the acoustic startle response (lowest effective dose 0.66 mg/kg). In addition, rolipram (0.66 mg/kg) blocks the disruptive effects of amphetamine (10 mg/kg) on PPI. At a slightly higher dose (1.0 mg/kg), rolipram also induces catalepsy. Thus, phosphodiesterase-4 (PDE4) inhibition has many of the same behavioral effects as traditional antipsychotic medications. In contrast to traditional antipsychotics, these effects are achieved through alteration of an intracellular second messenger system rather than antagonism of neurotransmitter receptors. Given previous reports showing rolipram improves cognition, we conclude that PDE4 represents an important novel target for further antipsychotic drug development.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Antipsychotic Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rolipram/pharmacology , Acoustic Stimulation , Animals , Behavior, Animal/drug effects , Catalepsy/chemically induced , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dextroamphetamine/antagonists & inhibitors , Dextroamphetamine/pharmacology , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/psychology , Haloperidol/pharmacology , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Reflex, Startle/drug effects
19.
Neuroscience ; 144(4): 1314-23, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17184927

ABSTRACT

Cigarette smoking is significantly more prevalent in individuals with schizophrenia than in non-affected populations. Certain neurocognitive deficits and disruptions common in schizophrenia may be altered by smoking, leading to the hypothesis that schizophrenics engage in smoking behavior to alleviate specific neurocognitive symptoms of the disorder. Additionally, research suggests that individuals with schizophrenia have altered auditory event-related potentials (ERPs) and abnormalities in evoked gamma oscillations which are both indices of sensory information processing. This study was conducted to examine the effect of acute administration of nicotine and the non-specific nicotinic antagonist mecamylamine on the P20 and N40 components of the ERP and evoked gamma oscillations in mice. Acute nicotine (1 mg/kg) significantly increased P20 amplitude, an effect that was blocked by pretreatment with mecamylamine (2 mg/kg). Additionally, acute nicotine increased the normal burst of evoked gamma following an auditory stimulus. The increase in evoked gamma was also blocked by mecamylamine pretreatment. Although acute nicotine decreased amplitude of the N40 component, this decrease was not attenuated by mecamylamine. These results replicate findings that nicotine may enhance early sensory information processing through the nicotinic acetylcholinergic receptor system in an established model (ERPs) and extend these findings in an emerging, novel model (evoked gamma oscillations) of sensory information processing. The results also support the hypothesis that nicotine may be beneficial to individuals with deficits in neurocognitive functions, such as those suffering from schizophrenia.


Subject(s)
Brain/drug effects , Evoked Potentials, Auditory/drug effects , Mecamylamine/pharmacology , Nicotine/pharmacology , Acetylcholine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biological Clocks/drug effects , Biological Clocks/physiology , Brain/metabolism , Brain/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Evoked Potentials, Auditory/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/physiopathology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tobacco Use Disorder/etiology , Tobacco Use Disorder/physiopathology
20.
Neuroscience ; 141(3): 1257-64, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16750890

ABSTRACT

People with schizophrenia display sensory encoding deficits across a broad range of electrophysiological and behavioral measures, suggesting fundamental impairments in the ability to transduce the external environment into coherent neural representations. This inability to create basic components of complex stimuli interferes with a high fidelity representation of the world and likely contributes to cognitive deficits. The current study evaluates the effects of constitutive forebrain activation of the G(s)alpha G-protein subunit on auditory threshold and gain using acoustic brainstem responses and cortically generated N40 event-related potentials to assess the role of cyclic AMP signaling in sensory encoding. Additionally, we examine the ability of pharmacological treatments that mimic (amphetamine) or ameliorate (haloperidol) positive symptoms of schizophrenia to test the hypothesis that the encoding deficits observed in G(s)alpha transgenic mice can be normalized with treatment. We find that G(s)alpha transgenic mice have decreased amplitude of cortically generated N40 but normal acoustic brainstem response amplitude, consistent with forebrain transgene expression and a schizophrenia endophenotype. Transgenic mice also display decreased stimulus intensity response (gain) in both acoustic brainstem response and N40, indicating corticofugal influence on regions that lack transgene expression. N40 deficits in transgenic animals were ameliorated with low dose haloperidol and reversed with higher dose, suggesting dopamine D2 receptor-linked Gi activity contributes to the impairment. Consistent with this hypothesis, we recreated the G(s)alpha transgenic deficit in wild type animals using the indirect dopamine agonist amphetamine. This transgenic model of sensory encoding deficits provides a foundation for identifying biochemical contributions to sensory processing impairments associated with schizophrenia.


Subject(s)
Auditory Threshold/physiology , Brain Stem/physiopathology , Cerebral Cortex/physiopathology , Evoked Potentials, Auditory/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Schizophrenia/physiopathology , Acoustic Stimulation/methods , Amphetamine/pharmacology , Animals , Auditory Threshold/drug effects , Auditory Threshold/radiation effects , Brain Stem/drug effects , Brain Stem/radiation effects , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/drug effects , Disease Models, Animal , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electroencephalography/methods , Evoked Potentials, Auditory/drug effects , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Haloperidol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Schizophrenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...