Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Semin Cell Dev Biol ; 129: 31-39, 2022 09.
Article in English | MEDLINE | ID: mdl-33975755

ABSTRACT

Olfactory dysfunction is manifested in a wide range of neurological and psychiatric diseases, and often emerges prior to the onset of more classical symptoms and signs. From a behavioral perspective, olfactory deficits typically arise in conjunction with impairments of cognition, motivation, memory, and emotion. However, a conceptual framework for explaining the impact of olfactory processing on higher brain functions in health and disease remains lacking. Here we aim to provide circuit-level insights into this question by synthesizing recent advances in olfactory network connectivity with other cortical brain regions such as the prefrontal cortex. We will focus on social cognition as a representative model for exploring and critically evaluating the relationship between olfactory cortices and higher-order cortical regions in rodent models. Although rodents do not recapitulate all dimensions of human social cognition, they have experimentally accessible neural circuits and well-established behavioral tests for social motivation, memory/recognition, and hierarchy, which can be extrapolated to other species including humans. In particular, the medial prefrontal cortex (mPFC) has been recognized as a key brain region in mediating social cognition in both rodents and humans. This review will highlight the underappreciated connectivity, both anatomical and functional, between the olfactory system and mPFC circuitry, which together provide a neural substrate for olfactory modulation of social cognition and social behaviors. We will provide future perspectives on the functional investigation of the olfactory-mPFC circuit in rodent models and discuss how to translate such animal research to human studies.


Subject(s)
Prefrontal Cortex , Social Cognition , Animals , Brain , Cognition , Humans , Social Behavior
2.
Mol Psychiatry ; 27(3): 1805-1815, 2022 03.
Article in English | MEDLINE | ID: mdl-35165396

ABSTRACT

Sensorimotor information processing underlies normal cognitive and behavioral traits and has classically been evaluated through prepulse inhibition (PPI) of a startle reflex. PPI is a behavioral dimension deregulated in several neurological and psychiatric disorders, yet the mechanisms underlying the cross-diagnostic nature of PPI deficits across these conditions remain to be understood. To identify circuitry mechanisms for PPI, we performed circuitry recording over the prefrontal cortex and striatum, two brain regions previously implicated in PPI, using wild-type (WT) mice compared to Disc1-locus-impairment (LI) mice, a model representing neuropsychiatric conditions. We demonstrated that the corticostriatal projection regulates neurophysiological responses during the PPI testing in WT, whereas these circuitry responses were disrupted in Disc1-LI mice. Because our biochemical analyses revealed attenuated brain-derived neurotrophic factor (Bdnf) transport along the corticostriatal circuit in Disc1-LI mice, we investigated the potential role of Bdnf in this circuitry for regulation of PPI. Virus-mediated delivery of Bdnf into the striatum rescued PPI deficits in Disc1-LI mice. Pharmacologically augmenting Bdnf transport by chronic lithium administration, partly via phosphorylation of Huntingtin (Htt) serine-421 and its integration into the motor machinery, restored striatal Bdnf levels and rescued PPI deficits in Disc1-LI mice. Furthermore, reducing the cortical Bdnf expression negated this rescuing effect of lithium, confirming the key role of Bdnf in lithium-mediated PPI rescuing. Collectively, the data suggest that striatal Bdnf supply, collaboratively regulated by Htt and Disc1 along the corticostriatal circuit, is involved in sensorimotor gating, highlighting the utility of dimensional approach in investigating pathophysiological mechanisms across neuropsychiatric disorders.


Subject(s)
Brain-Derived Neurotrophic Factor , Corpus Striatum , Nerve Tissue Proteins , Prefrontal Cortex , Prepulse Inhibition , Animals , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/metabolism , Humans , Mice , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/metabolism , Prepulse Inhibition/physiology , Reflex, Startle/physiology , Sensory Gating/physiology
3.
Proc Natl Acad Sci U S A ; 110(30): 12462-7, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23840059

ABSTRACT

Guided by features of molecular, cellular, and circuit dysfunction affecting the prefrontal cortex in clinical investigations, we targeted prefrontal cortex in studies of a model for neuropsychiatric illness using transgenic mice expressing a putative dominant-negative disrupted in schizophrenia 1 (DN-DISC1). We detected marked augmentation of GAPDH-seven in absentia homolog Siah protein binding in the DISC1 mice, a major hallmark of a nuclear GAPDH cascade that is activated in response to oxidative stress. Furthermore, deficits were observed in well-defined tests for the cognitive control of adaptive behavior using reversal learning and reinforcer devaluation paradigms. These deficits occurred even though DN-DISC1 mice showed intact performance in simple associative learning and normal responses in consumption of reward. In an additional series of assessments, motivational functions also were impoverished in DN-DISC1 mice, including tests of the dynamic modulation of reward value by effortful action, progressive ratio performance, and social behavior. Augmentation of an oxidative stress-associated cascade (e.g., a nuclear GAPDH cascade) points to an underlying condition that may contribute to the profile of cognitive and motivational impairments in DN-DISC1 mice by affecting the functional integrity of the prefrontal cortex and dysfunction within its connected networks. As such, this model should be useful for further preclinical research and drug discovery efforts relevant to the burden of prefrontal dysfunction in neuropsychiatric illness.


Subject(s)
Cognition Disorders/metabolism , Mental Disorders/metabolism , Motivation , Oxidative Stress , Prefrontal Cortex/metabolism , Animals , Mice , Mice, Inbred C57BL , Prefrontal Cortex/pathology , Social Behavior
4.
Neurobiol Dis ; 82: 176-184, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26093170

ABSTRACT

Cannabis is an increasingly popular and controversial drug used worldwide. Cannabis use often begins during adolescence, a highly susceptible period for environmental stimuli to alter functional and structural organization of the developing brain. Given that adolescence is a critical time for the emergence of mental illnesses before full-onset in early adulthood, it is particularly important to investigate how genetic insults and adolescent cannabis exposure interact to affect brain development and function. Here we show for the first time that a perturbation in disrupted in schizophrenia 1 (DISC1) exacerbates the response to adolescent exposure to delta-9-tetrahydrocannabinol (Δ(9)-THC), a major psychoactive ingredient of cannabis, consistent with the concept that gene-environment interaction may contribute to the pathophysiology of psychiatric conditions. We found that chronic adolescent treatment with Δ(9)-THC exacerbates deficits in fear-associated memory in adult mice that express a putative dominant-negative mutant of DISC1 (DN-DISC1). Synaptic expression of cannabinoid receptor 1 (CB1R) is down-regulated in the prefrontal cortex, hippocampus, and amygdala, critical brain regions for fear-associated memory, by either expression of DN-DISC1 or adolescent Δ(9)-THC treatment. Notably, elevation of c-Fos expression evoked by context-dependent fear memory retrieval is impaired in these brain regions in DN-DISC1 mice. We also found a synergistic reduction of c-Fos expression induced by cue-dependent fear memory retrieval in DN-DISC1 with adolescent Δ(9)-THC exposure. These results suggest that alteration of CB1R-mediated signaling in DN-DISC1 mice may underlie susceptibility to detrimental effects of adolescent cannabis exposure on adult behaviors.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Emotions/drug effects , Gene-Environment Interaction , Memory/drug effects , Nerve Tissue Proteins/genetics , Animals , Brain/metabolism , Disease Models, Animal , Emotions/physiology , Fear/drug effects , Fear/physiology , Memory/physiology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Cannabinoid, CB1/metabolism
5.
Hum Mol Genet ; 22(8): 1574-80, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23314019

ABSTRACT

Imaging of the human brain has been an invaluable aid in understanding neuropsychopharmacology and, in particular, the role of dopamine in the striatum in mental illness. Here, we report a study in a genetic mouse model for major mental illness guided by results from human brain imaging: a systematic study using small animal positron emission tomography (PET), autoradiography, microdialysis and molecular biology in a putative dominant-negative mutant DISC1 transgenic model. This mouse model showed augmented binding of radioligands to the dopamine D2 receptor (D2R) in the striatum as well as neurochemical and behavioral changes to methamphetamine administration. Previously we reported that this model displayed deficits in the forced swim test, a representative indicator of antidepressant efficacy. By combining the results of our two studies, we propose a working hypothesis for future studies that this model might represent a mixed condition of depression and psychosis. We hope that this study will also help bridge a major gap in translational psychiatry between basic characterization of animal models and clinico-pharmacological assessment of patients mainly through PET imaging.


Subject(s)
Dopamine/metabolism , Molecular Imaging , Nerve Tissue Proteins/genetics , Positron-Emission Tomography/methods , Receptors, Dopamine D2/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Mapping , Corpus Striatum/metabolism , Corpus Striatum/ultrastructure , Dopamine/genetics , Humans , Methamphetamine/administration & dosage , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Protein Binding , Radiography , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/isolation & purification
6.
Neuron ; 111(2): 220-235.e9, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36379214

ABSTRACT

Schizophrenia (SZ) and bipolar disorder (BP) are highly heritable major psychiatric disorders that share a substantial portion of genetic risk as well as their clinical manifestations. This raises a fundamental question of whether, and how, common neurobiological pathways translate their shared polygenic risks into shared clinical manifestations. This study shows the miR-124-3p-AMPAR pathway as a key common neurobiological mediator that connects polygenic risks with behavioral changes shared between these two psychotic disorders. We discovered the upregulation of miR-124-3p in neuronal cells and the postmortem prefrontal cortex from both SZ and BP patients. Intriguingly, the upregulation is associated with the polygenic risks shared between these two disorders. Seeking mechanistic dissection, we generated a mouse model that upregulates miR-124-3p in the medial prefrontal cortex. We demonstrated that the upregulation of miR-124-3p increases GRIA2-lacking calcium-permeable AMPARs and perturbs AMPAR-mediated excitatory synaptic transmission, leading to deficits in the behavioral dimensions shared between SZ and BP.


Subject(s)
Bipolar Disorder , MicroRNAs , Schizophrenia , Mice , Animals , Schizophrenia/genetics , Schizophrenia/metabolism , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Multifactorial Inheritance , Prefrontal Cortex/metabolism
7.
Neurobiol Dis ; 45(1): 48-56, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21914480

ABSTRACT

The molecular mechanisms of major mental illnesses, such as schizophrenia and bipolar disorder, are unclear. To address this fundamental question, many groups have studied molecular expression profiles in postmortem brains and other tissues from patients compared with those from normal controls. Development of unbiased high-throughput approaches, such as microarray, RNA-seq, and proteomics, have supported and facilitated this endeavor. In addition to genes directly involved in neuron/glia signaling, especially those encoding for synaptic proteins, genes for metabolic cascades are differentially expressed in the brains of patients with schizophrenia and bipolar disorder, compared with those from normal controls in DNA microarray studies. Here we propose the importance and usefulness of genetic mouse models in which such differentially expressed molecules are modulated. These animal models allow us to dissect the mechanisms of how such molecular changes in patient brains may play a role in neuronal circuitries and overall behavioral phenotypes. We also point out that models in which the metabolic genes are modified are obviously untested from mental illness viewpoints, suggesting the potential to re-address these models with behavioral assays and neurochemical assessments.


Subject(s)
Bipolar Disorder/genetics , Brain/metabolism , Schizophrenia/genetics , Animals , Bipolar Disorder/metabolism , Bipolar Disorder/physiopathology , Brain/physiopathology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Schizophrenia/metabolism , Schizophrenia/physiopathology
8.
Drug Metab Dispos ; 40(11): 2067-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22837388

ABSTRACT

D-Amino acid oxidase (DAAO) catalyzes the oxidative deamination of D-amino acids including D-serine, a full agonist at the glycine modulatory site of the N-methyl-d-aspartate (NMDA) receptor. To evaluate the significance of DAAO-mediated metabolism in the pharmacokinetics of oral D-serine, plasma D-serine levels were measured in both wild-type mice and transgenic mice lacking DAAO. Although D-serine levels were rapidly diminished in wild-type mice (t(½) = 1.2 h), sustained drug levels over the course of 4 h (t(½) > 10 h) were observed in mice lacking DAAO. Coadministration of D-serine with 6-chlorobenzo[d]isoxazol-3-ol (CBIO), a small-molecule DAAO inhibitor, in wild-type mice resulted in the enhancement of plasma D-serine levels, although CBIO seems to have only temporary effects on the plasma D-serine levels due to glucuronidation of the key hydroxyl group. These findings highlight the predominant role of DAAO in the clearance of D-serine from the systemic circulation. Thus, a potent DAAO inhibitor with a longer half-life should be capable of maintaining high plasma D-serine levels over a sustained period of time and might have therapeutic implications for the treatment of schizophrenia.


Subject(s)
D-Amino-Acid Oxidase/deficiency , D-Amino-Acid Oxidase/metabolism , Serine/pharmacokinetics , Animals , Brain/metabolism , D-Amino-Acid Oxidase/antagonists & inhibitors , D-Amino-Acid Oxidase/genetics , Female , Half-Life , Humans , Isoxazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microsomes, Liver/metabolism , Schizophrenia/blood , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/metabolism , Serine/blood , Serine/pharmacology
9.
Transl Psychiatry ; 12(1): 99, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35273151

ABSTRACT

Under the hypothesis that olfactory neural epithelium gene expression profiles may be useful to look for disease-relevant neuronal signatures, we examined microarray gene expression in olfactory neuronal cells and underscored Notch-JAG pathway molecules in association with schizophrenia (SZ). The microarray profiling study underscored JAG1 as the most promising candidate. Combined with further validation with real-time PCR, downregulation of NOTCH1 was statistically significant. Accordingly, we reverse-translated the significant finding from a surrogate tissue for neurons, and studied the behavioral profile of Notch1+/- mice. We found a specific impairment in social novelty recognition, whereas other behaviors, such as sociability, novel object recognition and olfaction of social odors, were normal. This social novelty recognition deficit was male-specific and was rescued by rapamycin treatment. Based on the results from the animal model, we next tested whether patients with psychosis might have male-specific alterations in social cognition in association with the expression of NOTCH1 or JAG1. In our first episode psychosis cohort, we observed a specific correlation between the expression of JAG1 and a face processing measure only in male patients. The expression of JAG1 was not correlated with any other cognitive and symptomatic scales in all subjects. Together, although we acknowledge the pioneering and exploratory nature, the present work that combines both human and animal studies in a reciprocal manner suggests a novel role for the Notch-JAG pathway in a behavioral dimension(s) related to social cognition in psychotic disorders in a male-specific manner.


Subject(s)
Psychotic Disorders , Animals , Down-Regulation , Female , Humans , Male , Mice , Olfactory Mucosa
10.
Psychiatr Clin North Am ; 43(2): 263-274, 2020 06.
Article in English | MEDLINE | ID: mdl-32439021

ABSTRACT

The onset of schizophrenia is usually in late adolescence or early adulthood. However, accumulating evidence has suggested that the disease condition is an outcome of gene-environment interactions that act in neural development during early life and adolescence. Some children who later develop schizophrenia have early developmental and educational and social challenges. Some patients with schizophrenia have an abundance of nonspecific neurologic soft signs and minor physical anomalies. Adolescence is a sensitive period of increased neuronal plasticity. It is important to consider early detection and intervention from the prodromal stage to early disease to prevent its devastating long-term consequences.


Subject(s)
Schizophrenia/diagnosis , Schizophrenic Psychology , Adolescent , Early Diagnosis , Humans , Prodromal Symptoms , Risk Factors , Schizophrenia/physiopathology
11.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32029441

ABSTRACT

Altered cortical excitation-inhibition (E-I) balance resulting from abnormal parvalbumin interneuron (PV IN) function is a proposed pathophysiological mechanism of schizophrenia and other major psychiatric disorders. Preclinical studies have indicated that disrupted-in-schizophrenia-1 (Disc1) is a useful molecular lead to address the biology of prefrontal cortex (PFC)-dependent cognition and PV IN function. To date, PFC inhibitory circuit function has not been investigated in depth in Disc1 locus impairment (LI) mouse models. Therefore, we used a Disc1 LI mouse model to investigate E-I balance in medial PFC (mPFC) circuits. We found that inhibition onto layer 2/3 excitatory pyramidal neurons in the mPFC was significantly reduced in Disc1 LI mice. This reduced inhibition was accompanied by decreased GABA release from local PV, but not somatostatin (SOM) INs, and by impaired feedforward inhibition (FFI) in the mediodorsal thalamus (MD) to mPFC circuit. Our mechanistic findings of abnormal PV IN function in a Disc1 LI model provide insight into biology that may be relevant to neuropsychiatric disorders including schizophrenia.


Subject(s)
Interneurons , Parvalbumins , Animals , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Somatostatin
12.
Article in English | MEDLINE | ID: mdl-30597182

ABSTRACT

Dopamine D2 receptor (D2R) hyperactivity causes altered brain development and later produces onset of symptoms mimicking schizophrenia. It is known that D2R interacts with disrupted in schizophrenia 1 (DISC1); however, the effect of D2R-DISC1 interaction in intracellular signalling and neurite growth has not been studied. This study investigated the effect of D2R over-activation on Akt-GSK3ß signalling and neurite morphology in cortical neurons. Over-activation of D2Rs caused neurite lesions, which were associated with decreased protein kinase B (Akt) and glycogen synthase kinase 3 beta (GSK3ß) phosphorylation in cortical neurons. The antipsychotic drug aripiprazole was more effective in the prevention of neurite lesions than haloperidol. Unlike haloperidol, aripiprazole prevented downregulation of phospho (p) Akt-pGSK3ß induced by D2R hyperactivity, indicating involvement of different pathways. D2Rs were hyperactive in cortical neurons of mice with DISC1 mutation, which caused more severe neurite lesions in cortical neurons treated with quinpirole. Immunofluorescent staining for Ca2+/calmodulin-dependent protein kinase II (CaMKII) confirmed that cortical pyramidal neurons were involved in the D2R hyperactivity-induced neurite lesions. Using the fluorescence resonance energy transfer (FRET) technique, we provide direct evidence that D2R hyperactivity led to D2R-DISC1 complex formation, which altered pGSK3ß signalling. This study showed that D2R hyperactivity-induced D2R-DISC1 complex formation is associated with decreased pAkt-pGSK3ß signalling and in turn, caused neurite impairment. Aripiprazole and haloperidol prevented the impairment of neurite growth but appeared to do so via different intracellular signalling pathways.


Subject(s)
Aripiprazole/pharmacology , Haloperidol/pharmacology , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Neuroprotective Agents/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurites/metabolism , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/physiology , Neuroprotection/drug effects , Neuroprotection/physiology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics
13.
Schizophr Res ; 210: 149-156, 2019 08.
Article in English | MEDLINE | ID: mdl-31204062

ABSTRACT

DISC1 was originally expected to be a genetic risk factor for schizophrenia, but the genome wide association studies have not supported this idea. In contrast, neurobiological studies of DISC1 in cell and animal models have demonstrated that direct perturbation of DISC1 protein elicits neurobiological and behavioral abnormalities relevant to a wide range of psychiatric conditions, in particular psychosis. Thus, the utility of DISC1 as a biological lead for psychosis research is clear. In the present study, we aimed to capture changes in the molecular landscape in the prefrontal cortex upon perturbation of DISC1, using the Disc1 locus impairment (Disc1-LI) model in which the majority of Disc1 isoforms have been depleted, and to explore potential molecular mediators relevant to psychiatric conditions. We observed a robust change in gene expression profile elicited by Disc1-LI in which the stronger effects on molecular networks were observed in early stage compared with those in adulthood. Significant alterations were found in specific pathways relevant to psychiatric conditions, such as pathways of signaling by G protein-coupled receptor, neurotransmitter release cycle, and voltage gated potassium channels. The differentially expressed genes (DEGs) between Disc1-LI and wild-type mice are significantly enriched not only in neurons, but also in astrocytes and oligodendrocyte precursor cells. The brain-disorder-associated genes at the mRNA and protein levels rather than those at the genomic levels are enriched in the DEGs. Together, our present study supports the utility of Disc1-LI mice in biological research for psychiatric disorder-associated molecular networks.


Subject(s)
Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Prefrontal Cortex/metabolism , Schizophrenia/genetics , Transcriptome , Age Factors , Animals , Disease Models, Animal , Genetic Loci , Male , Mice , Mice, Transgenic , Sequence Analysis, RNA
14.
J Cereb Blood Flow Metab ; 39(7): 1306-1313, 2019 07.
Article in English | MEDLINE | ID: mdl-29430995

ABSTRACT

Although still a matter of controversy, disrupted in schizophrenia protein 1 (DISC1) was suggested as a potential inhibitor of phosphodiesterase 4 (PDE4). We used Disc1 locus impairment (LI) mice to investigate the interaction between PDE4 and DISC 1 in vivo and in vitro. [11C](R)-Rolipram binding was measured by PET in LI (n = 11) and C57BL/6 wild-type (WT, n = 9) mice. [11C](R)-Rolipram total distribution volumes (VT) were calculated and corrected for plasma-free fraction (fP) measured in a separate group of LI (n = 6) and WT (n = 7) mice. PDE4 enzyme activity was measured using in vitro samples of cerebral cortices from groups of LI (n = 4), heterozygote (n = 4), and WT (n = 4) mice. Disc1 LI mice showed a 41% increase in VT (18 ± 6 vs. 13±4 mL/cm3, P = 0.04) compared to WT mice. VT/fP showed a 73% significant increase (90 ± 31 vs. 52 ± 15 mL/cm3, P = 0.004) in Disc1 LI compared to WT mice. PDE4 enzymatic activity assay confirmed in vivo findings showing significant group differences (p < 0.0001). In conclusion, PDE4 activity was increased in the absence of critical DISC1 protein isoforms both in vivo and in vitro. Additionally, [11C](R)-Rolipram PET was sensitive enough to assess altered PDE4 activity caused by PDE4-DISC1 interaction.


Subject(s)
Cerebral Cortex/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Nerve Tissue Proteins/metabolism , Positron-Emission Tomography/methods , Animals , Carbon Radioisotopes , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Deletion , Haploinsufficiency , Heterozygote , Homozygote , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Phosphodiesterase 4 Inhibitors , Rolipram/metabolism
15.
Mol Neuropsychiatry ; 4(1): 20-29, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29998115

ABSTRACT

A truncated disrupted in schizophrenia 1 (Disc1) gene increases the risk of psychiatric disorders, probably affecting cortical interneurons. Here, we sought to determine whether this cell population is affected in mice carrying a truncated (Disc1) allele (DN-DISC1). We utilized whole cell recordings to assess electrophysiological properties and modulation by dopamine (DA) in two classes of interneurons: fast-spiking (FS) and low threshold-spiking (LTS) interneurons in wild-type and DN-DISC1 mice. In DN-DISC1 mice, FS interneurons, but not LTS interneurons, exhibited altered action potentials. Further, the perineuronal nets that surround FS interneurons exhibited abnormal morphology in DN-DISC1 mice, and the DA modulation of this cell type was altered in DN-DISC1 mice. We conclude that early-life manipulation of a gene associated with risk of psychiatric disease can result in dysfunction, but not loss, of specific GABAergic interneurons. The resulting alteration of excitatory-inhibitory balance is a critical element in DISC1 pathophysiology.

16.
Neurosci Res ; 105: 75-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26458529

ABSTRACT

Chemokines play important roles in the central nervous system, including mediating neuroinflammation and guiding the intracortical migration of interneurons during development. Alteration in parvalbumin-positive interneurons is a key neuropathological hallmark of multiple mental conditions. We recently reported a significant reduction in the expression of CXCL12 in olfactory neurons from sporadic cases with schizophrenia compared with matched controls, suggesting a role for CXCR4/CXCL12 signaling in mental conditions. Thus, we depleted the chemokine receptor Cxcr4 from mice using the parvalbumin-2A-Cre line. The conditional knockout mice exhibited a unique behavioral phenotype involving increased stereotypy. Stereotypy is observed in many psychiatric conditions, including schizophrenia, autism, and dementia. Thus, the Cxcr4 conditional knockout mice may serve as a model for this symptomatic feature.


Subject(s)
Receptors, CXCR4/genetics , Stereotyped Behavior , Animals , Male , Maze Learning , Mice, Knockout , Neurons/metabolism , Parvalbumins/metabolism , Reflex, Startle
17.
Neurosci Res ; 112: 63-69, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27354230

ABSTRACT

Sleep disturbances are common in psychiatric disorders, but the causal relationship between the two and the underlying genetic factors is unclear. The DISC1 gene is strongly linked to mood disorders and schizophrenia in a Scottish pedigree. In an earlier study we found a sleep homeostasis disturbance in a Drosophila model overexpressing wild-type human DISC1. Here we aimed to explore the relationship between sleep and the DISC1 gene in a mammalian model, a novel transgenic mouse model expressing full-length human DISC1. We assessed circadian rhythms by monitoring wheel running activity under normal 24-h light:dark conditions and in constant darkness and found the DISC1 mice to have normal circadian photoentrainment and normal intrinsic circadian period. We also assessed sleep duration and quality in the DISC1 mice and found that they were awake longer than wild-type controls at baseline with a tendency for lower rebound of delta activity during recovery from a short sleep deprivation. Thus we suggest that DISC1 may be involved in sleep regulation.


Subject(s)
Nerve Tissue Proteins/metabolism , Sleep/physiology , Animals , Circadian Rhythm , Humans , Mice, Transgenic , Nerve Tissue Proteins/genetics , Wakefulness/physiology
18.
Mol Neuropsychiatry ; 2(1): 28-36, 2016 May.
Article in English | MEDLINE | ID: mdl-27606318

ABSTRACT

Despite the recent progress in psychiatric genetics, very few studies have focused on genetic risk factors in glial cells that, compared to neurons, can manifest different molecular pathologies underlying psychiatric disorders. In order to address this issue, we studied the effects of mutant disrupted in schizophrenia 1 (DISC1), a genetic risk factor for schizophrenia, in cultured primary neurons and astrocytes using an unbiased mass spectrometry-based proteomic approach. We found that selective expression of mutant DISC1 in neurons affects a wide variety of proteins predominantly involved in neuronal development (e.g., SOX1) and vesicular transport (Rab proteins), whereas selective expression of mutant DISC1 in astrocytes produces changes in the levels of mitochondrial (GDPM), nuclear (TMM43) and cell adhesion (ECM2) proteins. The present study demonstrates that DISC1 variants can perturb distinct molecular pathways in a cell type-specific fashion to contribute to psychiatric disorders through heterogenic effects in diverse brain cells.

19.
Mol Neuropsychiatry ; 1(1): 52-59, 2015 May.
Article in English | MEDLINE | ID: mdl-26417572

ABSTRACT

We present results from a novel comparative approach to the study of mechanisms of psychiatric disease. Previous work examined neural activity patterns in the hippocampus of a freely behaving mouse model associated with schizophrenia, the calcineurin knockout mouse. Here we examined a genetically distinct mouse that exhibits a similar set of behavioral phenotypes associated with schizophrenia, a transgenic model expressing a putative dominant-negative DISC1 (DN-DISC1). Strikingly, the principal finding of the earlier work is replicated in the DN-DISC1 mice, that is, a selective increase in the numbers of sharp-wave ripple events in the local hippocampal LFP, while at the same time other LFP patterns such as theta and gamma are unaffected. Sharp-wave ripples are thought to arise from hippocampal circuits, and reflect the coordinated activity of the principal excitatory cells of the hippocampus, in specific patterns that represent reactivated memories of previous experiences and imagined future experiences that predict behavior. These findings suggest that multiple genetic alterations could converge on distinct patterns of aberrant neurophysiological function to give rise to common behavioral phenotypes in psychiatric disease.

20.
Neurosci Res ; 98: 45-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25697395

ABSTRACT

The pericentriolar material (PCM) is composed of proteins responsible for microtubule nucleation/anchoring at the centrosome, some of which have been associated with genetic susceptibility to schizophrenia. Here, we show that mice haploinsufficient for Pericentriolar material 1 (Pcm1(+/-)), which encodes a component of the PCM found to bear rare loss of function mutations in patients with psychiatric illness, manifest neuroanatomical phenotypes and behavioral abnormalities. Using ex vivo magnetic resonance imaging of the Pcm1(+/-) brain, we detect reduced whole brain volume. Pcm1 mutant mice show impairment in social interaction, specifically in the social novelty phase, but not in the sociability phase of the three-chamber social interaction test. In contrast, Pcm1(+/-) mice show normal preference for a novel object, suggesting specific impairment in response to novel social stimulus. In addition, Pcm1(+/-) mice display significantly reduced rearing activity in the open field. Pcm1(+/-) mice behave normally in the elevated plus maze, rotarod, prepulse inhibition, and progressive ratio tests. Together, our results suggest that haploinsufficiency at the Pcm1 locus can induce a range of neuroanatomical and behavioral phenotypes that support the candidacy of this locus in neuropsychiatric disorders.


Subject(s)
Brain/pathology , Cell Cycle Proteins/genetics , Mental Disorders/genetics , Animals , Exploratory Behavior , Haploinsufficiency , Male , Mental Disorders/pathology , Mental Disorders/psychology , Mice, Mutant Strains , Motor Activity , Organ Size , Prepulse Inhibition , Reflex, Startle , Schizophrenia/genetics , Social Behavior
SELECTION OF CITATIONS
SEARCH DETAIL