Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Psychiatry ; 11: 713, 2020.
Article in English | MEDLINE | ID: mdl-32793004

ABSTRACT

The past 5 years have seen a sharp increase in the number of studies using calcium imaging in behaving rodents. These studies have helped identify important roles for individual cells, brain regions, and circuits in some of the core behavioral phenotypes of psychiatric disorders, such as schizophrenia and autism, and have characterized network dysfunction in well-established models of these disorders. Since rescuing clinically relevant behavioral deficits in disease model mice remains a foundation of preclinical CNS research, these studies have the potential to inform new therapeutic approaches targeting specific cell types or projections, or perhaps most importantly, the network-level context in which neurons function. In this mini-review, we will provide a brief overview of recent insights into psychiatric disease-associated mouse models and behavior paradigms, focusing on those achieved by cellular resolution imaging of calcium dynamics in neural populations. We will then discuss how these experiments can support efforts within the pharmaceutical industry, such as target identification, assay development, and candidate screening and validation. Calcium imaging is uniquely capable of bridging the gap between two of the key resources that currently enable CNS drug discovery: genomic and transcriptomic data from human patients, and translatable, population-resolution measures of brain activity (such as fMRI and EEG). Applying this knowledge could yield real value to patients in the near future.

2.
Front Syst Neurosci ; 13: 45, 2019.
Article in English | MEDLINE | ID: mdl-31551721

ABSTRACT

The primary auditory cortex processes acoustic sequences for the perception of behaviorally meaningful sounds such as speech. Sound information arrives at its input layer four from where activity propagates to associative layer 2/3. It is currently not known whether there is a characteristic organization of neuronal population activity across layers and sound levels during sound processing. Here, we identify neuronal avalanches, which in theory and experiments have been shown to maximize dynamic range and optimize information transfer within and across networks, in primary auditory cortex. We used in vivo 2-photon imaging of pyramidal neurons in cortical layers L4 and L2/3 of mouse A1 to characterize the populations of neurons that were active spontaneously, i.e., in the absence of a sound stimulus, and those recruited by single-frequency tonal stimuli at different sound levels. Single-frequency sounds recruited neurons of widely ranging frequency selectivity in both layers. We defined neuronal ensembles as neurons being active within or during successive temporal windows at the temporal resolution of our imaging. For both layers, neuronal ensembles were highly variable in size during spontaneous activity as well as during sound presentation. Ensemble sizes distributed according to power laws, the hallmark of neuronal avalanches, and were similar across sound levels. Avalanches activated by sound were composed of neurons with diverse tuning preference, yet with selectivity independent of avalanche size. Our results suggest that optimization principles identified for avalanches guide population activity in L4 and L2/3 of auditory cortex during and in-between stimulus processing.

3.
Transl Psychiatry ; 8(1): 3, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29317600

ABSTRACT

Disturbed activity patterns in cortical networks contribute to the pathophysiology of schizophrenia (SZ). Several lines of evidence implicate NMDA receptor hypofunction in SZ, and blocking NMDA receptor signaling during early neurodevelopment produces cognitive deficits in rodent models that resemble those seen in schizophrenic patients. However, the altered network dynamics underlying these cognitive impairments largely remain to be characterized, especially at the cellular level. Here, we use in vivo two-photon calcium imaging to describe pathological dynamics, occurring in parallel with cognitive dysfunction, in a developmental NMDA receptor hypofunction model. We observed increased synchrony and specific alterations in spatiotemporal activity propagation, which could be causally linked to a previously unidentified persistent bursting phenotype. This phenotype was rescued by acute treatment with the NMDA receptor co-agonist D-serine or the GABAB receptor agonist baclofen, which similarly rescued working memory performance. It was not reproduced by optogenetic inhibition of fast-spiking interneurons. These results provide novel insight into network-level abnormalities mediating the cognitive impairment induced by NMDA receptor hypofunction.


Subject(s)
Cognitive Dysfunction/metabolism , GABA-B Receptor Agonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/metabolism , Animals , Cognitive Dysfunction/chemically induced , Excitatory Amino Acid Antagonists/toxicity , Female , Interneurons/metabolism , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Rats , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Signal Transduction/drug effects
4.
Nat Commun ; 6: 10118, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26656849

ABSTRACT

Neuregulin-1 (NRG1) and its receptor ErbB4 influence several processes of neurodevelopment, but the mechanisms regulating this signalling in the mature brain are not well known. DISC1 is a multifunctional scaffold protein that mediates many cellular processes. Here we present a functional relationship between DISC1 and NRG1-ErbB4 signalling in mature cortical interneurons. By cell type-specific gene modulation in vitro and in vivo including in a mutant DISC1 mouse model, we demonstrate that DISC1 inhibits NRG1-induced ErbB4 activation and signalling. This effect is likely mediated by competitive inhibition of binding of ErbB4 to PSD95. Finally, we show that interneuronal DISC1 affects NRG1-ErbB4-mediated phenotypes in the fast spiking interneuron-pyramidal neuron circuit. Post-mortem brain analyses and some genetic studies have reported interneuronal deficits and involvement of the DISC1, NRG1 and ErbB4 genes in schizophrenia, respectively. Our results suggest a mechanism by which cross-talk between DISC1 and NRG1-ErbB4 signalling may contribute to these deficits.


Subject(s)
Interneurons/physiology , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-4/metabolism , Synapses/physiology , Animals , Cells, Cultured , Cerebral Cortex , Humans , Mice , Nerve Tissue Proteins/genetics , Neuregulin-1/genetics , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4/genetics , Signal Transduction/physiology
5.
Elife ; 4: e07224, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26151674

ABSTRACT

Spontaneous fluctuations in neuronal activity emerge at many spatial and temporal scales in cortex. Population measures found these fluctuations to organize as scale-invariant neuronal avalanches, suggesting cortical dynamics to be critical. Macroscopic dynamics, though, depend on physiological states and are ambiguous as to their cellular composition, spatiotemporal origin, and contributions from synaptic input or action potential (AP) output. Here, we study spontaneous firing in pyramidal neurons (PNs) from rat superficial cortical layers in vivo and in vitro using 2-photon imaging. As the animal transitions from the anesthetized to awake state, spontaneous single neuron firing increases in irregularity and assembles into scale-invariant avalanches at the group level. In vitro spike avalanches emerged naturally yet required balanced excitation and inhibition. This demonstrates that neuronal avalanches are linked to the global physiological state of wakefulness and that cortical resting activity organizes as avalanches from firing of local PN groups to global population activity.


Subject(s)
Action Potentials , Cerebral Cortex/physiology , Pyramidal Cells/physiology , Wakefulness , Animals , Cerebral Cortex/cytology , Image Processing, Computer-Assisted , Microscopy, Fluorescence, Multiphoton , Models, Neurological , Rats
6.
Neurobiol Dis ; 53: 26-35, 2013 May.
Article in English | MEDLINE | ID: mdl-23336981

ABSTRACT

Schizophrenia (SZ) is often described as a disease of neuronal connectivity. Cognitive processes such as working memory, which are particularly dependent on the proper functioning of complex cortical circuitry, are disturbed in the disease. Reciprocal connections between pyramidal neurons and interneurons, as well as dopaminergic innervations, form the basis for higher cognition in the cortex. Nonetheless, only a few review articles are available which address how each synapse operates, and is possibly disturbed in SZ, at least in part by the mechanisms involving genetic susceptibility factors for SZ. In this review, we provide an overview of cortical glutamatergic, GABAergic, and dopaminergic circuitry, review SZ-associated deficits at each of these synapses, and discuss how genetic factors for SZ may contribute to SZ-related phenotype deficits in a synapse-specific manner. Pinpointing the spatially and temporally distinct sites of action of putative SZ susceptibility factors may help us better understand the pathological mechanisms of SZ, especially those associated with synaptic functioning and neuronal connectivity.


Subject(s)
Cerebral Cortex/pathology , Nerve Net/physiopathology , Schizophrenia/pathology , Synapses/pathology , Animals , Cerebral Cortex/physiopathology , Humans , Nerve Net/metabolism , Nerve Net/pathology , Neurons/metabolism , Neurons/pathology , Neurons/physiology , Presynaptic Terminals/pathology , Presynaptic Terminals/physiology , Schizophrenia/physiopathology , Synapses/physiology
7.
Science ; 339(6117): 335-9, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23329051

ABSTRACT

Environmental stressors during childhood and adolescence influence postnatal brain maturation and human behavioral patterns in adulthood. Accordingly, excess stressors result in adult-onset neuropsychiatric disorders. We describe an underlying mechanism in which glucocorticoids link adolescent stressors to epigenetic controls in neurons. In a mouse model of this phenomenon, a mild isolation stress affects the mesocortical projection of dopaminergic neurons in which DNA hypermethylation of the tyrosine hydroxylase gene is elicited, but only when combined with a relevant genetic risk for neuropsychiatric disorders. These molecular changes are associated with several neurochemical and behavioral deficits that occur in this mouse model, all of which are blocked by a glucocorticoid receptor antagonist. The biology and phenotypes of the mouse models resemble those of psychotic depression, a common and debilitating psychiatric disease.


Subject(s)
Adolescent Behavior , Adolescent Development , Affective Disorders, Psychotic/metabolism , Dopaminergic Neurons/metabolism , Epigenesis, Genetic , Glucocorticoids/metabolism , Stress, Psychological/metabolism , Adolescent , Affective Disorders, Psychotic/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Stress, Psychological/genetics
8.
Nature ; 473(7345): 92-6, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21471969

ABSTRACT

Regulatory mechanisms governing the sequence from progenitor cell proliferation to neuronal migration during corticogenesis are poorly understood. Here we report that phosphorylation of DISC1, a major susceptibility factor for several mental disorders, acts as a molecular switch from maintaining proliferation of mitotic progenitor cells to activating migration of postmitotic neurons in mice. Unphosphorylated DISC1 regulates canonical Wnt signalling via an interaction with GSK3ß, whereas specific phosphorylation at serine 710 (S710) triggers the recruitment of Bardet-Biedl syndrome (BBS) proteins to the centrosome. In support of this model, loss of BBS1 leads to defects in migration, but not proliferation, whereas DISC1 knockdown leads to deficits in both. A phospho-dead mutant can only rescue proliferation, whereas a phospho-mimic mutant rescues exclusively migration defects. These data highlight a dual role for DISC1 in corticogenesis and indicate that phosphorylation of this protein at S710 activates a key developmental switch.


Subject(s)
Cerebral Cortex/embryology , Nerve Tissue Proteins , Neurons/cytology , Neurons/physiology , Stem Cells/cytology , Animals , COS Cells , Cell Movement/genetics , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Chlorocebus aethiops , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , PC12 Cells , Phosphorylation , Protein Binding , Rats , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism
9.
Proc Natl Acad Sci U S A ; 108(14): 5795-800, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436042

ABSTRACT

NMDA receptors are key regulators of synaptic plasticity, and their hypofunction is thought to contribute to the pathophysiology of CNS disorders. Furthermore, NMDA receptors participate in the formation, maintenance, and elimination of synapses. The consequences of NMDA receptor hypofunction on synapse biology were explored in a genetic mouse model, in which the levels of NMDA receptors are reduced to 10% of normal levels (i.e., NR1-knockdown mice). In these mice, synapse number is reduced in an age-dependent manner; reductions are observed at the postpubertal age of 6 wk, but normal at 2 wk of age. Efforts to uncover the biochemical underpinnings of this phenomenon reveal synapse-specific reductions in 14-3-3ε protein and in Disrupted in Schizophrenia-1 (DISC1), two schizophrenia susceptibility factors that have been implicated in the regulation of spine density. Subchronic administration of MK-801, an NMDA receptor antagonist, produces similar synaptic reductions in both spine density and DISC1, indicating that synaptic levels of DISC1 are regulated by NMDA receptor function. The synaptic reduction of DISC1 and 14-3-3ε is developmentally correlated with the age-dependent decrease in striatal spine density.


Subject(s)
Corpus Striatum/cytology , Dendritic Spines/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , 14-3-3 Proteins/metabolism , Age Factors , Analysis of Variance , Animals , Blotting, Western , Corpus Striatum/physiology , Dendritic Spines/metabolism , Dizocilpine Maleate/pharmacology , Electrophoresis, Gel, Two-Dimensional , Fluorescent Antibody Technique , Gene Knockdown Techniques , Immunohistochemistry , Locomotion/physiology , Mice , Microscopy, Electron , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Social Behavior
10.
Biochem Biophys Res Commun ; 400(4): 631-7, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20807500

ABSTRACT

Disrupted-in-Schizophrenia 1 (DISC1) is a promising genetic risk factor for major mental disorders. Many groups repeatedly reported a role for DISC1 in brain development in various strains of mice and rats by using RNA interference (RNAi) approach. Nonetheless, due to the complexity of its molecular disposition, such as many splice variants and a spontaneous deletion in a coding exon of the DISC1 gene in some mouse strains, there have been debates on the interpretation on these published data. Thus, in this study, we address this question by DISC1 knockdown via short-hairpin RNAs (shRNAs) against several distinct target sequences with more than one delivery methodologies into several mouse strains, including C57BL/6, ICR, and 129X1/SvJ. Here, we show that DISC1 knockdown by in utero electroporation of shRNA against exons 2, 6, and 10 consistently results in neuronal migration defects in the developing cerebral cortex, which are successfully rescued by co-expression of full-length DISC1. Furthermore, lentivirus-mediated shRNA also led to migration defects, which is consistent with two other methodologies already published, such as plasmid-mediated and retrovirus-mediated ones. The previous study by Song's group also reported that, in the adult hippocampus, the phenotype elicited by DISC1 knockdown with shRNA targeting exon 2 was consistently seen in both C57BL/6 and 129S6 mice. Taken together, we propose that some of DISC1 isoforms that are feasible to be knocked down by shRNAs to exon 2, 6, and 10 of the DISC1 gene play a key role for neuronal migration commonly in various mouse strains and rats.


Subject(s)
Cell Movement/genetics , Cerebral Cortex/embryology , Nerve Tissue Proteins/genetics , Neurons/physiology , RNA Interference , Animals , Cerebral Cortex/cytology , Electroporation/methods , Female , Gene Knockdown Techniques , Gene Transfer Techniques , Lentivirus , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Rats , Uterus
11.
Proc Natl Acad Sci U S A ; 107(12): 5622-7, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212127

ABSTRACT

Neuregulin-1 (NRG1) and Disrupted-in-Schizophrenia-1 (DISC1) are promising susceptibility factors for schizophrenia. Both are multifunctional proteins with roles in a variety of neurodevelopmental processes, including progenitor cell proliferation, migration, and differentiation. Here, we provide evidence linking these factors together in a single pathway, which is mediated by ErbB receptors and PI3K/Akt. We show that signaling by NRG1 and NRG2, but not NRG3, increase expression of an isoform of DISC1 in vitro. Receptors ErbB2 and ErbB3, but not ErbB4, are responsible for transducing this effect, and PI3K/Akt signaling is also required. In NRG1 knockout mice, this DISC1 isoform is selectively reduced during neurodevelopment. Furthermore, a similar decrease in DISC1 expression is seen in beta-site amyloid precursor protein cleaving enzyme-1 (BACE1) knockout mice, in which NRG1/Akt signaling is reportedly impaired. In contrast to neuronal DISC1 that was reported and characterized, expression of DISC1 in other types of cells in the brain has not been addressed. Here we demonstrate that DISC1, like NRG and ErbB proteins, is expressed in neurons, astrocytes, oligodendrocytes, microglia, and radial progenitors. These findings may connect NRG1, ErbBs, Akt, and DISC1 in a common pathway, which may regulate neurodevelopment and contribute to susceptibility to schizophrenia.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Astrocytes/metabolism , Brain/metabolism , Cell Line , Cells, Cultured , Humans , Mice , Mice, Knockout , Microglia/metabolism , Nerve Tissue Proteins/genetics , Neuregulin-1/deficiency , Neuregulin-1/genetics , Neurogenesis , Neurons/metabolism , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schizophrenia/etiology , Signal Transduction
12.
Neuron ; 65(4): 480-9, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20188653

ABSTRACT

Adult brain function and behavior are influenced by neuronal network formation during development. Genetic susceptibility factors for adult psychiatric illnesses, such as Neuregulin-1 and Disrupted-in-Schizophrenia-1 (DISC1), influence adult high brain functions, including cognition and information processing. These factors have roles during neurodevelopment and are likely to cooperate, forming pathways or "signalosomes." Here we report the potential to generate an animal model via in utero gene transfer in order to address an important question of how nonlethal deficits in early development may affect postnatal brain maturation and high brain functions in adulthood, which are impaired in various psychiatric illnesses such as schizophrenia. We show that transient knockdown of DISC1 in the pre- and perinatal stages, specifically in a lineage of pyramidal neurons mainly in the prefrontal cortex, leads to selective abnormalities in postnatal mesocortical dopaminergic maturation and behavioral abnormalities associated with disturbed cortical neurocircuitry after puberty.


Subject(s)
Behavior, Animal/physiology , Dopamine/metabolism , Frontal Lobe/metabolism , Nerve Tissue Proteins/genetics , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cell Differentiation/genetics , Cell Lineage/physiology , Chromatography, High Pressure Liquid , Clozapine/pharmacology , Dopamine Agents/pharmacology , Electrophysiology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Gene Transfer Techniques , Immunohistochemistry , Methamphetamine/pharmacology , Mice , Microdialysis , Motor Activity/drug effects , Motor Activity/genetics , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Interference , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Sensory Gating/drug effects , Sensory Gating/genetics , Spatial Behavior/drug effects , Spatial Behavior/physiology
13.
Nat Neurosci ; 13(3): 327-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139976

ABSTRACT

Synaptic spines are dynamic structures that regulate neuronal responsiveness and plasticity. We examined the role of the schizophrenia risk factor DISC1 in the maintenance of spine morphology and function. We found that DISC1 anchored Kalirin-7 (Kal-7), regulating access of Kal-7 to Rac1 and controlling the duration and intensity of Rac1 activation in response to NMDA receptor activation in both cortical cultures and rat brain in vivo. These results explain why Rac1 and its activator (Kal-7) serve as important mediators of spine enlargement and why constitutive Rac1 activation decreases spine size. This mechanism likely underlies disturbances in glutamatergic neurotransmission that have been frequently reported in schizophrenia that can lead to alteration of dendritic spines with consequential major pathological changes in brain function. Furthermore, the concept of a signalosome involving disease-associated factors, such as DISC1 and glutamate, may well contribute to the multifactorial and polygenetic characteristics of schizophrenia.


Subject(s)
Dendritic Spines/physiology , Glutamic Acid/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Synapses/physiology , rac1 GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Excitatory Postsynaptic Potentials/physiology , Gene Knockdown Techniques , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Time Factors , Tissue Culture Techniques
14.
Trends Neurosci ; 32(9): 485-95, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19712980

ABSTRACT

Schizophrenia (SZ) is primarily an adult psychiatric disorder in which disturbances caused by susceptibility genes and environmental insults during early neurodevelopment initiate neurophysiological changes over a long time course, culminating in the onset of full-blown disease nearly two decades later. Aberrant postnatal brain maturation is an essential mechanism underlying the disease. Currently, symptoms of SZ are treated with anti-psychotic medications that have variable efficacy and severe side effects. There has been much interest in the prodromal phase and the possibility of preventing SZ by interfering with the aberrant postnatal brain maturation associated with this disorder. Thus, it is crucial to understand the mechanisms that underlie the long-term progression to full disease manifestation to identify the best targets and approaches towards this goal. We believe that studies of certain SZ genetic susceptibility factors with neurodevelopmental implications will be key tools in this task. Accumulating evidence suggests that neuregulin-1 (NRG1) and disrupted-in-schizophrenia-1 (DISC1) are probably functionally convergent and play key roles in brain development. We provide an update on the role of these emerging concepts in understanding the complex time course of SZ from early neurodevelopmental disturbances to later onset and suggest ways of testing these in the future.


Subject(s)
Brain/growth & development , Brain/metabolism , ErbB Receptors/metabolism , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Schizophrenia/pathology , Animals , Disease Models, Animal , ErbB Receptors/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Nerve Tissue Proteins/genetics , Receptor, ErbB-4 , Schizophrenia/metabolism
15.
Proc Natl Acad Sci U S A ; 104(36): 14501-6, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17675407

ABSTRACT

Here, we report generation and characterization of Disrupted-In-Schizophrenia-1 (DISC1) genetically engineered mice as a potential model for major mental illnesses, such as schizophrenia. DISC1 is a promising genetic risk factor for major mental illnesses. In this transgenic model, a dominant-negative form of DISC1 (DN-DISC1) is expressed under the alphaCaMKII promoter. In vivo MRI of the DN-DISC1 mice detected enlarged lateral ventricles particularly on the left side, suggesting a link to the asymmetrical change in anatomy found in brains of patients with schizophrenia. Furthermore, selective reduction in the immunoreactivity of parvalbumin in the cortex, a marker for an interneuron deficit that may underlie cortical asynchrony, is observed in the DN-DISC1 mice. These results suggest that these transgenic mice may be used as a model for schizophrenia. DN-DISC1 mice also display several behavioral abnormalities, including hyperactivity, disturbance in sensorimotor gating and olfactory-associated behavior, and an anhedonia/depression-like deficit.


Subject(s)
Disease Models, Animal , Genes, Dominant/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/genetics , Schizophrenia/physiopathology , Animals , Behavior, Animal , Biomarkers , Humans , Mice , Mice, Transgenic , Phenotype , Schizophrenia/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...